<<

Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 DOI 10.1186/s40478-017-0468-y

REVIEW Open Access RNA of disease-associated repeat expansions Kushal J. Rohilla1 and Keith T. Gagnon1,2*

Abstract , or simple sequences, occur naturally in the human and have important roles in genome evolution and function. However, the expansion of microsatellites is associated with over two dozen neurological diseases. A common denominator among the majority of these disorders is the expression of expanded tandem repeat-containing RNA, referred to as xtrRNA in this review, which can mediate molecular disease pathology in multiple ways. This review focuses on the potential impact that simple tandem repeat expansions can have on the biology and of RNA that contain them and underscores important gaps in understanding. Merging the of repeat expansion disorders with the current understanding of RNA biology, including splicing, , transport, turnover and , will help clarify mechanisms of disease and improve therapeutic development. Keywords: Repeat expansion disease, Microsatellite, Tandem repeats, RNA, Splicing, Transcription, Transport, Export, Turnover, Translation, Mechanism, Therapeutics, Myotonic , DM1, DM2, Huntington's disease, HD, C9ORF72, C9FTD/ALS, Amyotrophic lateral sclerosis, , SCA, Fragile X, SBMA, FXTAS

Introduction understanding of the underlying molecular biology and In 1991 it was discovered that a microsatellite sequence pathology remain, which in turn limits identification of expansion is the cause of two distinct neurological disor- promising therapeutic approaches. The goal of this re- ders, (FXS) [303] and spinal bulbar view is to help address these gaps by discussing the po- muscular (SBMA), or Kennedy's disease [167]. tential impact of xtrRNA on cellular RNA metabolism. Since then, simple repeat sequence expansions have We begin with an overview that covers microsatellite been associated with over twenty more neurological dis- origin, evolution, and expansion. We then follow orders [166, 300, 333] (Table 1). What has been learned xtrRNA through its cycle, beginning with transcrip- is that microsatellite expansions may cause disease in tion and continuing through splicing, folding, in- multiple ways. For nearly all of these neurological disor- teractions, localization, turnover, and translation. We ders, however, disease includes production of RNA that rationalize the logic of current molecular disease models, contains the aberrant repeat expansion sequence. Ac- note where important mechanistic information is lack- cordingly, the leading disease mechanisms involve repeat ing, and emphasize new pathways to consider for mech- expansion RNA-mediated sequestration of critical RNA- anistic insight. We use this discussion to also highlight binding and translation of repeat expansion areas where therapeutic intervention may be useful. RNA into toxic repetitive polypeptides. Tremendous progress has been made in understanding the metabolism of expanded tandem repeat-containing Origin and expansion of microsatellites in human RNA (xtrRNA). Nonetheless, various gaps in our disease Simple tandem repeat sequences in the Microsatellite sequences comprise approximately 3% of * Correspondence: [email protected] the human genome, about twice as much as protein cod- 1 Department of and Molecular Biology, Southern Illinois ing sequence [1, 171]. Microsatellites, interchangeably University, School of Medicine, Carbondale, Illinois, USA 2Department of Chemistry and Biochemistry, Southern Illinois University, known as simple or short tandem repeats (STRs), are Carbondale, Illinois, USA

© The Author(s). 2017 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 2 of 22

Table 1 Microsatellite repeat expansion disorders Disorder Repeating Genomic Normal Pathogenic Expanded repeats result in: Repeat discovery & unit location name length length references Gene xtrRNA xtrRNA xtrRNA silencing transcription proteins foci FXS/FRAXA† CGG 5’UTR FMR1 6-55 200+ Yes* No No No [114, 161, 230, 303] SBMA† CAG Coding AR 9-36 38-62 No Yes Yes* L .D. [167] DM1 CTG 3’UTR DMPK 5-37 50-10000 No Yes* Yes Yes* [193] HD CAG Coding HTT 10-35 35+ No Yes Yes* L.D. [192] SCA1 CAG Coding ATXN1 6-35 49-88 No Yes Yes* L.D. [232] FRAXE CCG 5'UTR AFF2 4-39 200-900 Yes* No No No [152] DRPLA CAG Coding ATN1 6-35 49-88 No Yes Yes* L.D. [155] SCA3 CAG Coding ATXN3 12-40 55-86 No Yes Yes* Yes [140] SCA2 CAG Coding ATXN2 14-32 33-77 No Yes Yes* L.D. [127, 253] FRDA GAA FXN 8-33 90+ Yes* Yes / No No No [30] SCA6 CAG Coding CACNA1A 4-18 21-30 No Yes Yes* L.D. [337] EPM1 CCCCGC CSTB 2-3 30-80 Yes* Yes / No No No [170] CCCGCG SCA7 CAG Coding ATXN7 7-17 38-120 No Yes Yes* L.D. [57] OPMD GCG Coding PABPN1 6-10 12-17 No Yes Yes* No [23] SCA8 CTG 3’UTR ATXN8 16-34 74+ No Yes Yes* Yes* [156] SCA12 CAG 5’UTR PPP2R2B 7-28 66-78 No Yes* No No [121] SCA10 ATTCT Intron ATXN10 10-20 500-4500 No Yes ? Yes* [205] SCA17 CAG Coding TBP 25-42 47-63 No Yes Yes* L.D. [224] DM2 CCTG Intron CNBP 10-26 75-11000 No Yes ? Yes* [184] FXTAS/FXPOI CGG 5’UTR FMR1 6-55 55-200 No Yes Yes* Yes* [143, 287] HDL2 CTG/CAG 3'UTR/antisense JPH3 <50 50+ No Yes ? Yes* [120] SCA31 TGGAA Intron TK2/BEAN 0 45+ No Yes Yes* Yes* [256] SCA36 GGCCTG Intron Nop56 3-14 650+ No Yes ? Yes* [153] C9FTD/ALS GGGGCC Intron C9ORF72 2-25 25+ No Yes Yes* Yes* [59, 248] FRA7A CGG Intron ZFN713 5-22 85+ No Yes* / No No No [209] FRA2A CGG Intron AFF3 8-17 300+ No Yes* / No No No [210] Disorders are listed in order of the year they were discovered, with the appropriate references relating to their discovery. This table highlights known RNA biology for each disease with respect to xtrRNA transcription, translation, and formation of nuclear focal aggregates Dagger symbol (†) indicates that athough the CAG repeat for SBMA was discovered first, the CGG repeat for FXS was published first. Asterisk (*) indicates the most likely repeat-associated disease mechanism(s) for that disorder. L.D. length-dependent, SBMA Spinal-Bulbar Muscular Atrophy, EPM1 Progressive Myoclonus Epilepsy 1 (Unverricht–Lundborg Disease), FXS/FRAXA Fragile X Syndrome, DM Myotonic Dystrophy, HD Huntington’s Disease, SCA Spinocerebellar Ataxia, FRAXE Fragile X E Syndrome, DRPLA Dentatorubral-Pallidoluysian Atrophy, FRDA Friedreich Ataxia, OPMD Oculopharyngeal , FXTAS Fragile X–Associated Tremor/Ataxia Syndrome, FXPOI Fragile X-Associated Primary Ovarian Insufficiency, HDL2 Huntington’sDisease-Like2,C9FTD/ALS C9ORF72-Associated and Amyotrophic Lateral Sclerosis, FRA7A CGG Expansion at Fragile Site 7A, FRA2A CGG Expansion at Fragile Site 2A usually defined as simple sequence motifs of one to six The origin and evolution of microsatellites is incom- that are contiguously repeated at least a few pletely understood. They may have derived from simple times [24, 69]. Microsatellites occur throughout the gen- and repetitive sequence motifs found in mobile genetic ome, but are predominantly found in noncoding pro- elements, such as non-LTR () retro- moters, , 5' and 3' untranslated regions (UTRs), transposons like Alu and L1 [6, 51, 95]. Transposable el- and intergenic regions [236, 257, 291]. Intergenic micro- ements have colonized the human genome extensively satellites seem to fit neutral evolution models, although and their remains have undergone and replica- not perfectly [69], and are among the most variable gen- tion, providing the starting material for simple tandem omic sequences [32]. Therefore, they serve as the basis repeats [69]. The dense repetitive sequence of centro- for forensic DNA analyses and as markers for population meres and is proposed to originate from in- studies [24, 61, 65, 236]. corporation of during early Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 3 of 22

eukaryotic evolution [87, 304]. Small sequence duplica- damage. Multiple DNA damage control pathways have tions of these simple sequences can further produce been implicated, including mechanisms that replace microsatellites with multiple repeats. The STRs that are DNA bases, like base excision repair (BER) or expanded in Friedreich ataxia (FRDA) and myotonic excision repair (NER), especially as sources for repeat dystrophy type 2 (DM2), for example, have been traced expansion in non-dividing cells [206]. However, mis- to an origin [41, 163]. In contrast, de novo match repair (MMR) has been argued to be a primary genesis by events like random mutation, replication slip- driver of repeat expansion [75, 106, 130, 260, 271]. page, and duplication of unique sequence may also ac- MMR expands repeats through recognition and process- count for the birth of microsatellite sequences [28, 69]. ing of unusual DNA structures, such as small bulges and STRs have been shown to have positive roles in evolu- hairpins [260], via the enzyme MutSβ (MSH2-MSH3 tion, such as bacterial resistance to and circa- complex) [130, 260, 334]. The processing and damage dian clock adaptation to the environment in Neurospora rectification steps are carried out by MutSβ and associ- crassa and Drosophila melanogaster [83, 133, 211, 258, ated proteins, including the MutLα (MLH1-PMS2 com- 279, 307]. The placement of microsatellite sequences in plex) or MutLγ (MLH1-MLH3 complex) and near regulatory and coding regions of the genome that help remove DNA lesions [106, 130, 241]. Polymer- also implicate them in control of and ases like Polβ are then recruited, which can insert extra genetic interactions [133, 331]. repeats due to flawed priming or templating [33, 190]. An important question is how repeats are able to ex- Expansion of simple tandem repeats pand out of control, sometimes into the hundreds or There are several distinct mechanisms that can contrib- thousands of perfect tandem copies, without accumulat- ute to the expansion of naturally occurring microsatel- ing significant interruptions? Microsatellites that are lites. In this section we provide a brief overview of these evolutionarily neutral, typically in intergenic regions, be- mechanisms. Many excellent reviews on this topic are come highly mutable when they exceed thresholds above cited in this section and recommended for further read- just a few tandem repeats [68, 95, 320]. Therefore, the ing (see [84, 130, 149, 206, 212, 260, 297, 331, 333]). likelihood of remaining as a perfect tandem repeat with- A major source of microsatellite expansion in dividing out interruption is expected to decrease with tandem re- cells is DNA replication, although mitotic recombination peat length. This suggests that accumulation of large is also recognized as a contributing factor [84, 149, 212, expansions must either occur quickly, before 242]. During replication, repetitive sequences can cause can accumulate, or their disruption must be guarded problems at the replication fork and result in fork rever- against [320]. Genic regions of the genome, where all cur- sals or template switching, which can insert extra re- rently known disease-associated repeat expansions occur peats [76, 144, 149, 212]. At the strand level, polymerase [31, 236] (Table 1), seem to enjoy special favor through slipping can cause expansions in the leading or lagging positive evolutionary selection processes that protect se- strand [84, 137, 144, 149]. Repeats may also induce im- quence fidelity [191, 236, 284]. However, it seems unlikely perfect Okazaki fragment ligation and add repeats in the that this would contribute significantly to large repeat lagging strand [81, 93, 278]. The pathway followed for expansions. For example, non-repetitive codons would expansion of a repeat has been proposed to be a balan- presumably be preferred and selected over unstable repeat cing act between several factors [84, 149, 212]. These in- codons. clude relative repeat length, the stability and types of Mechanisms have been proposed that could provide non-canonical structures the repeat sequence can form, large expansions in a single step, including template and nearby flanking sequences. After repeat sequences switching replication models where repeats are already are added to one or both strands, the daughter strands sufficiently large enough [225, 266] and out-of-register reanneal. Misalignment and slippage will occur and extra synthesis during -based re- sequences will bulge out to form non-canonical (non-B- pair of double-strand breaks (DSBs) [212, 242, 249, 250, form) structures like hairpins or quadruplexes [237, 331]. 283]. One intriguing mechanism for rapid and large re- If these structures persist to the next round of replication, peat accumulation is break-induced replication (BIR) or if they undergo flawed repair, they can result in per- [148, 176]. BIR is a homologous recombination pathway manent expansions [130, 149, 212, 260, 297]. During that can rescue collapsed or broken replication forks DNA recombination, which repairs single-end or double- [195]. It is induced when a replisome collides with a strand breaks, unequal crossing over or template switch- broken single-end DSB [189]. BIR is also believed to be ing can cause misalignments and introduction of add- selective for structure-prone or GC-rich repeats that are itional repeats [208, 242, 306]. long enough to form stable structures [148]. In this Repeat expansion events are intimately tied to the re- mechanism of expansion, stable structures would cause pair of non-canonical DNA structures and DNA fork reversals. Resolution of these four-way junction Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 4 of 22

structures would result in a one-ended DSB. To restart (FRA7A) and fragile 2A (FRA2A) fragile site loci [209, 210]. the fork, the one-ended DSB invades the sister chromatid Comparing and contrasting these disorders can highlight to form a D-loop, but likely does so out-of-register be- several trends. Almost half of the microsatellite expansion cause of the repetitive sequence, thus leading to expan- disorders result from CAG trinucleotide expansions, sion. While this BIR study was performed in , the mostly occurring in coding . All STRs for known re- results are expected to translate to human cells [176]. peat expansion disorders are GC-rich except for the trinu- Incremental expansions, such as those caused by cleotide GAA repeat of FRDA and the ATTCT and MMR, are typically on the order of 1-3 repeats at a time TGGAA pentanucleotide repeats of spinocerebellar ataxia [260]. Could these events generate large uninterrupted 10 (SCA10) and 31 (SCA31), respectively. In this review expansions? Rapid accumulation of expansions via MMR we focus on large microsatellite repeat expansions that or other DNA damage repair pathways might be facili- are transcribed into RNA, a feature that is shared by tated by transcription across the repeat. It has been nearly all repeat expansion disorders (Table 1). shown that transcription is required for expansion of the Microsatellite expansions cause disease through two CGG repeat in a mouse model of FXS [2, 333]. Several broad molecular mechanisms (Fig. 1): loss-of-function studies have shown that transcription at repeat expan- for the associated gene or gain-of-function for the repeat sions is associated with repeat instability, possibly via expansion sequence. In loss of function mechanisms, formation of DNA-RNA hybrids, or R-loops [180, 181, gene expression can be silenced at the transcriptional 183, 195, 223, 246, 333]. It is possible that these events level, such as by epigenetic modification, resulting in the could allow DNA damage. One report has shown a complete loss of that gene's normal functions [70, 112]. correlation between R-loop formation and replication Alternatively, the affected gene may lose function at the fork stalling, offering a familiar mechanism for repeat protein level by the introduction of unusually long poly- expansion through DNA replication [86, 109]. Alterna- peptide tracts in the translated protein product (Fig. 1) tive mechanisms might involve oxidation of free DNA [168, 268]. In gain-of-function mechanisms the repetitive strands, or simple misalignment upon strand reanneal- polypeptide can take on new roles, such as protein ag- ing, to signal DNA damage repair [180]. The latter gregation. Many of these mutant misfolded proteins can- model suggests that GC-rich or structure-prone repeats not be degraded efficiently and will accumulate in would be more susceptible to expansion during tran- cellular aggregates or inclusions [48, 168, 332]. Aggrega- scription, which might explain why transcription levels tion also tends to sequester proteins and critical cellular alone are not predictive of expansion [333]. Thus, cycles components and is taxing on cellular proteostasis [48]. of transcription and R-loop formation might accelerate The xtrRNA can also acquire gain of function mecha- repeat expansion for structure-prone repeats via ongoing nisms, primarily through interaction with - DNA damage repair [180]. A -based model where binding proteins (Fig. 1). The repetitive xtrRNA forms transcription levels or R-loop formation could be con- length-dependent focal aggregates in cell nuclei in trolled, repeat sequence and size altered, expansions several diseases [35, 59, 196, 262, 311]. Loss-of- monitored, and DNA damage repair mechanisms sys- function and gain-of-function mechanisms can result tematically tested (perhaps building on HeLa cell models in complicated molecular disease pathologies and some recently described [187]) might allow more direct testing disorders can simultaneously exhibit multiple mecha- of these ideas. nisms (Table 1). A common theme among sources of repeat instability and expansion is DNA metabolism associated with Transcription and splicing at simple tandem strand separation and reannealing at microsatellite se- repeat expansions quences. These events can lead to formation of non- Transcribing repeat expansion sequences canonical structures and recruitment of DNA damage Repeat expansion sequences are known to inhibit or im- responses that ultimately and inadvertently add more re- pede RNA Polymerase II (Pol II) initiation or elongation peats. Thus, mechanisms meant to maintain and protect either directly or via induction of a repressed chromatin the genome can also lead to large tandem repeat expan- state [100]. Expansions like the GAA repeat in FRDA sions and cause human disease [11, 130, 333]. [19, 94, 97, 162, 231], the CTG repeat in myotonic dys- trophy type 1 (DM1) [25], the GGGGCC repeat in Microsatellite repeat expansion disorders C9ORF72-associated frontotemporal dementia and Since it was first discovered that microsatellite expan- amyotrophic lateral sclerosis (C9FTD/ALS) [108], and the sions can cause disease, at least two dozen microsatellite CGG repeat in FXS (also known as FRAXA) [44, 285] repeat expansion disorders have been subsequently re- have all been implicated in reduced or silenced transcrip- ported (Table 1). The latest discoveries are autism tion. For FXS [230, 298], Fragile XE (FRAXE) [18], FRDA spectrum disorders caused by expansions in fragile 7A [97], FRA2A [210] and FRA7A [209], transcription Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 5 of 22

Fig. 1 Distinct loss-of-function and gain-of-function mechanisms of disease for various repeat expansion disorders. Repeat expansions can occur in 5’ or 3’ UTRs, exons, or introns. Expanded tandem repeat-containing RNA (xtrRNA) may not be transcribed due to epigenetic silencing, thereby causing loss of gene function. If transcribed, xtrRNA may become trapped in the where it can form focal aggregates and functionally deplete important RNA binding proteins. The xtrRNA may also be exported to the where it can undergo translation to produce repeat- containing polypeptides that disrupt cellular processes. In some cases, xtrRNA can form focal nuclear aggregates and also be translated into repeat-containing polypeptides. Repeat-containing polypeptides can be toxic in multiple ways, including insoluble aggregation, blocking normal host protein function, inhibiting nucleocytoplasmic transport, and disrupting other critical cellular functions appears to be blocked or significantly reduced by DNA 2 (HDL2), among other diseases [26, 40, 126, 312]. Slo- of the repeat expansion or nearby CpG wed transcription across a repeat may also be able to in- islands. However, although transcription may be well duce antisense transcription of the non-template DNA below basal levels, it is possible that xtrRNA can still con- strand via R-loop formation [270]. For example, FRDA- tribute to disease in some cases [44]. associated GAA repeat expansion sequences were shown Slowed or stalled transcription across repeat expan- to initiate transcription and act as promoters in yeast sions may lead to R-loops, which further slow transcrip- [330]. However, many exhibit bidirectional tran- tion [123] and inadvertently contribute to deposition of scription [293] and in microsatellite diseases bidirec- repressive chromatin marks and silence transcription tional transcription typically initiates outside of the (Fig. 2) [44, 99, 316, 317]. R-loops play important roles repeat (Fig. 2) [26, 113]. Bidirectional transcription across in biology, such as immunoglobulin class switching repeats can also result in double R-loops that amplify re- [323], keeping CpG islands unmethylated [91, 254], and peat instability and accelerate methylation and transcrip- defining transcription termination signals [254, 270]. R- tional silencing [181, 183, 223]. Antisense transcription loop formation is common in transcription of C-rich can often interfere with transcription of the coding gene template sequences [324], which most disease-associated [145]. Most relevant to this review is the production of repeat expansion genomic loci possess. The impact of R- two xtrRNAs from bidirectional transcription and the po- loop formation on disease at repeat expansions is still tential to synthesize repetitive polypeptides from both unclear. Whether R-loop formation will trigger DNA xtrRNA. For example, in C9FTD/ALS both xtrRNAs form methylation, transcriptional silencing, or other events nuclear foci [59, 90, 248, 340] that sequester RNA-binding may be dependent upon a number of factors specific to proteins [47, 175, 217] and are translated into repetitive the affected gene or . polypeptides [9, 216], highlighting the importance of bidir- ectional transcription to molecular disease pathology. Bidirectional transcription of repeat expansions Bidirectional transcription has been reported to occur in The role of Supt4h in xtrRNA transcription DM1, C9FTD/ALS, Huntington's disease (HD), spino- Transcribing microsatellite expansions into xtrRNA re- cerebellar ataxia 8 (SCA8), and Huntington's disease-like quires processivity across repetitive sequence tracts that Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 6 of 22

Fig. 2 Effects of repeat expansion sequence on transcription. Repeat expansion sequences can perturb transcription by a epigenetic silencing, b inducing or facilitating bidirectional transcription, c reduced transcription kinetics, or d generating transcripts that can potentially be processed into small that could guide degradation or silencing of various complementary RNAs, including the xtrRNA itself can have very high GC content. The 5,6-dichloro-1-β-D- periods of extended pausing [50, 151, 309]. Long repeti- ribofuranosylbenzimidazole (DRB) sensitivity-inducing tive sequences prone to formation of secondary structure factor (DSIF), composed of Supt4h and Supt5h proteins in the transcription bubble, such as repeat-induced hair- (Spt4 and Spt5 in yeast), aids RNA Polymerase II (Pol II) pin or R-loop structures, may represent prime sites for in transcription elongation and transcription rate pausing or backtracking [251, 260, 333]. [305, 308]. The DSIF complex is important for tra- DSIF is also used by RNA Pol I to presumably ensure versing sequences that elicit pausing of RNA Pol II [305] robust transcription of abundant and repetitive riboso- and has been identified as a factor involved in the tran- mal RNA [122, 309]. It is worth noting that repeat ex- scription of RNA containing large simple repeat se- pansions might occur in ribosomal RNA genes but they quences. For example, transcription of repeat-containing have either not been characterized or have not been as- RNA from the huntingtin and C9ORF72 genes signifi- sociated with disease [122]. In contrast, RNA Pol III, cantly decreases when Supt4h is deleted or knocked- which only transcribes relatively small noncoding RNA down [159, 186]. Supt5h is a conserved transcription fac- genes, does not interact with the DSIF complex [309]. tor with a homolog known as NusG in that is Thus, transcription is unlikely to be successful if large important for elongation and processivity [185, 202]. microsatellite expansions occur in the small RNA genes Supt5h binds directly to the clamp coiled-coil domain of transcribed by RNA Pol III. These observations may RNA Pol II while Supt4h interacts through contact with lend some rationale as to why all disease-associated re- Supt5h [17, 119, 201]. Together, the DSIF complex inter- peat expansions to date are associated with Pol II- acts with the DNA template outside of the transcription transcribed genic regions [7, 31, 236]. bubble [17, 50, 151, 185]. Supt4h has a zinc-finger do- main that may be important for modulating DNA inter- Splicing of xtrRNA actions of DSIF [308], and thereby improve processivity Splicing involves several regulated steps, many accessory by maintaining RNA Pol II template interaction during factors and the spliceosome, a complex multi-component Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 7 of 22

enzyme. There is currently a lack of mechanistic insight interplay of modular protein and RNA interactions that regarding how the splicing apparatus reacts when encoun- are difficult to predict at present and local sequence and tering pre-mRNA containing large repetitive sequence context will likely be important for understanding the im- tracts [14]. Since introns can be excessively large while still pact of expanded repeats on splicing [14]. allowing productive and accurate splicing [263], the size of the repeat expansion itself is not expected to significantly Therapeutic approaches to control xtrRNA transcription impede splicing. However, transcription rates across and splicing microsatellite expansions can be reduced, which can influ- Characterizing the effect of microsatellite expansions on ence alternative splicing [58, 270], and stem loop struc- transcription and splicing will directly benefit thera- tures in large pre-mRNA introns have been predicted to peutic approaches for repeat expansion disorders. Proof- affect splicing [263]. of-principle methods to locally disrupt the interactions Examples of microsatellite repeat expansions modulat- of xtrRNA at repeat expansion loci, such as R-loops, ing splicing include the GAA repeat expansion associ- have been demonstrated for FXS and FRDA using small ated with FRDA. When placed near reporter gene exons and nucleic acids [44, 177]. Disrupting the or in the first intron of a minigene system, the interaction of Spt4 and Spt5, or modulating Spt4 func- GAA repeat caused complex splicing defects and accu- tion, could provide a therapeutic avenue for a number of mulation of aberrant splice products [15]. The mechan- repeat expansion disorders by reducing xtrRNA expres- ism proposed involved binding of various splicing sion. This has been demonstrated for CAG and factors to the GAA repeat-containing transcripts [15]. In GGGGCC repeat expansions [159, 186] and might be C9FTD/ALS, the intronic GGGGCC repeat has been particularly valuable in disorders exhibiting bidirectional implicated in splicing by favoring retention of the transcription across the repeat expansion. For splicing- intron-containing repeat, suggesting a mechanism by based therapeutics, blocking inclusion of repeat which C9ORF72 xtrRNA can escape to the cytoplasm expansion-containing introns, such as with splice- for translation [227]. Expanded CAG repeats of HD are modulating antisense or small RNAs, also linked to production of short alternatively spliced could prove to be useful for disorders like FRDA and forms of the huntingtin mRNA that contain the CAG re- C9FTD/ALS. peat expansion and add to the production of toxic poly- With the emergence of gene editing technologies, the glutamine protein [255]. direct removal of repeat expansions from the genome may also be possible. Removal of genomic repeat expan- Potential impact on splicing factors sions could eliminate the possibility of xtrRNA expres- If repeat expansion sequences can mimic the binding sion or reverse repressive epigenetic states. Careful SNP motif of splicing regulators, they could recruit splicing selection followed by targeting with CRISPR-Cas9 has factors and affect splice site selection. In DM1 the been shown to block promoter function and silence the MBNL family of splicing factors and CUG binding pro- mutant expanded in HD [215] or completely delete teins (CUGBPs) have an affinity for repetitive CUG and large portions of the mutant HD allele [265]. Targeting CAG sequence. Although the splicing of DM1 protein CRISPR-Cas9 to sequences flanking the CTG repeat in (DMPK) mRNA does not appear to be affected DM1 also caused large repeat deletions [299]. In model by the CUG repeat expansion that it contains, the spli- cells of DM1, CRISPR-Cas9 was used to introduce a cing pattern of an antisense transcript across the DMPK poly-A signal upstream of the CTG expansion in the repeat, which contains a CAG repeat, appears to be al- DMPK gene to prevent CUG repeat transcription, which tered by the expansion [105]. In HD the expanded CAG led to a reversal of molecular disease [318]. While poten- repeats have been proposed to interact with the splicing tial CRISPR-based therapeutics are exciting, precautions factor SRSF6, which is believed to contribute to altered must be taken to address potential pitfalls and challenges splicing to generate truncated repeat-containing hun- like off-target effects, delivery, and cell-type specific tingtin mRNA [255]. mechanisms of DNA damage repair [16, 54, 71, 85, 229, Repeat expansion sequences in xtrRNA could also 238, 240, 322]. alter splicing by recruiting factors that are not typically involved in splicing. These factors might modulate splice Structure, protein interactions, and localization of site selection or spliceosome activity by changing local xtrRNA ribonucleoprotein (RNP) structure or access to splice Structure of xtrRNAs and targeting with small molecules signals [67]. The repetitive structural nature of repeat During and after the synthesis and processing of expansions could also sterically hinder access to splice xtrRNA, the repetitive RNA will fold into repetitive and signals, depending on their proximity to splice enhancer unique structures and interact with proteins that have or silencer elements. Alternative splicing is a complicated an affinity for its sequence or structure. Watson-Crick Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 8 of 22

pairing apparently dominates folding since all atomic downstream from 22. MBNL-1 usually interacts resolution investigations of disease-associated xtrRNA to with these motifs to cause inclusion of exon 22 but in date, including CAG, CUG, CCG, CGG, CCUG, DM1 exon 22 is excluded during splicing [118, 34]. AUUCU, and CCCCGG, are imperfect A-form-like du- Blocking the interaction of proteins with DM1 xtrRNA plexes [39, 62, 147, 234, 341]. These structures possess by using oligonucleotides rescued splicing repeating units of Watson-Crick and mismatch paired defects and molecular pathology [310]. Thus, a major nucleotides [147, 341]. While some studies have identi- contributor to disease mechanism in DM1 is the se- fied G-quadruplexes or tetraplexes [45, 79, 194, 247], questration of splicing factors, particularly MBNL other reports suggest that xtrRNA either do not form proteins. quadruplexes or are transient and interconvert readily A number of diseases are characterized by binding of with Watson-Crick paired conformations, especially as specific proteins to xtrRNA or colocalization of proteins the number of repeating units increase [62, 108, 281, with xtrRNA focal aggregates (Table 1) [327]. These 329, 341]. Some reports of tetraplex structure may be include proteins like MBNL-1 in DM1, DM2, HD, the result of unusual interactions like dimerization be- spinocerebellar ataxia 3 (SCA3), SCA8 and HDL2 tween imperfect repeat duplex RNA, as was observed [197, 282, 327], hnRNP K in SCA10 and C9FTD/ALS for CGG repeat RNAs [103]. Convincing evidence for [46, 311], Pur-α, hnRNP F and SRSF2 in C9FTD/ALS the presence or biological significance of RNA G- [47, 108, 319], and Sam68 and hnRNP A2/B1 in quadruplexes inside human cells is still lacking [20, 107, FXTAS [262, 276]. As such, protein interactions with 164, 194], therefore direct roles for quadruplex RNA in xtrRNAplaykeyrolesindiseasemechanismandare repeat expansion disease remain unclear. expected to be important mediators of aberrant Available structures of short repeat RNAs reveal A- xtrRNA localization and aggregation [214]. Foci con- form-like conformations with unique mismatches that taining xtrRNA are believed to be the result of RNA- may be targeted with artificial molecules to selectively binding protein sequestration that can functionally bind repeat expansion RNA structure. Small deplete those proteins and partially protect the screening and structure-guided synthesis have experi- xtrRNA from degradation [214, 327]. mentally identified a variety of small molecules that can Sequence specific interactions may not entirely explain bind xtrRNA, such as the CUG, CCUG, CGG, and xtrRNA localization or foci formation. While certain GGGGCC repeat RNAs associated with DM1, DM2, proteins that prefer to bind G-rich sequence, like hnRNP FXS or FXTAS (Fragile X-associated tremor/ataxia syn- H/F, have been found to associate strongly with the drome), and C9FTD/ALS, respectively [38, 39, 226, 281, GGGGCC repeats of C9FTD/ALS, other interacting pro- 292, 314, 325]. These molecules have been shown to teins do not appear to have strong GGGGCC sequence- stabilize repetitive structure or disrupt protein binding, binding specificity, such as ALY/REF, SC-35, SF2, and which can correct molecular disease markers like nu- nucleolin [47, 108, 175]. Imperfect A-form-like duplexes, clear RNA foci and repetitive polypeptide translation, or or duplexes inter-converting with tetraplex conforma- improve pathology in cells and animal models. Although tions, may attract proteins that recognize the unique promising, their eventual therapeutic application will structures of xtrRNA rather than the specific sequence. need to demonstrate exquisite specificity for the RNA --rich (GAR) proteins containing RGG/ target, minimal non-specific interactions, and pharmaco- RG motifs, for example, are believed to recognize the logic safety and efficacy [102, 252]. structure of their nucleic acid partners rather than se- quence [289]. The GAR domain-containing proteins Protein interactions and localization of xtrRNA FUS (fused in sarcoma), FMRP, and hnRNP U all Both sequence specific and structure specific interac- recognize structured -rich RNA sequences with tions likely underlie protein binding to xtrRNA. The re- an apparent preference for transitions between canonical petitive nature of xtrRNA can result in multiple tandem duplexes and non-canonical structures like quadruplexes binding sites for proteins. In DM1, the disease- [233]. One explanation for foci is that proteins bind spe- associated xtrRNA contains hundreds or thousands of cifically to repeat sequence or structural elements of CUG repeats that bind and recruit possibly as many xtrRNA and then seed aggregation that recruits add- copies of MBNL-1 protein and potentially other CUG- itional secondary interacting factors. Thus, xtrRNA may binding proteins [173, 197, 235]. MBNL-1 recognizes CG form foci by either merging with existing nuclear bodies dinucleotides separated by 1-17 nucleotides [92], which or else establishing their own novel versions of RNA include motifs in pre-mRNA where MBNL-1 helps to granules. While focal aggregation of xtrRNA can be det- regulate splicing [245]. Examples include the pre-mRNA rimental to sequestered protein function, it may also of sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 1 protect the cell by preventing nuclear escape and trans- (SERCA1), which contains several YGCU(U/G)Y motifs lation of repeat RNAs [150]. Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 9 of 22

xtrRNA localization and membrane-free cellular organelles structures [21, 89, 179]. The disruption of membrane- Whether there is a specific localization pattern of free organelles, which are abundant in the nucleus, is xtrRNA inside cell nuclei is not entirely clear. Foci might linked to disease [198, 228, 272]. In fact, the disruption be expected to nucleate at the site of transcription. of membrane-free organelle assembly and dynamics by DMPK mRNA usually localizes to SC-35 splicing repetitive poly-glycine-arginine (poly-GR) and poly- speckles after transcription. However, when containing proline-arginine (poly-PR) translation products has CUG repeat expansions, the DMPK mRNA has been emerged as a leading molecular disease mechanism for shown to localize peri-transcriptionally outside of SC-35 C9FTD/ALS [165, 174, 182]. Association of certain splicing speckles [274]. RNA containing CAG, CUG and proteins with xtrRNA, dependent upon RNA sequence GGGGCC repeats were also shown to localize to SC-35 and structure, may strongly influence the subsequent splicing speckles and nuclear speckles [132, 295]. localization of xtrRNA with membrane-free cellular However, in other studies the xtrRNAs, specifically CUG compartments. and CGG RNAs, appeared to form foci stochastically [243, 280]. Live cell imaging of Spinach2 aptamer-tagged Abundance and turnover of xtrRNA CGG repeat xtrRNA revealed rapid aggregation and for- Abundance of foci-forming xtrRNA mation of very stable foci [280]. CGG xtrRNA foci were Understanding the biology of an RNA includes knowing additionally found to be mobile and dynamic and colo- the effective concentration or abundance of that RNA calized with Sam68 protein. They migrated around the and its turnover and decay pathways. Three current nucleus over time and could be seen to merge into lar- studies highlight the importance of characterizing cellu- ger foci or disaggregate into smaller foci. Live cell im- lar xtrRNA abundance. The cellular abundance of CUG aging of CUG repeat xtrRNA tagged with the MS2-GFP repeat-containing transcripts was recently measured system found similar effects for aggregation, foci forma- using transgenes and endogenous DMPK RNA in mouse tion and dynamics [243]. CUG repeat RNA foci forma- models of DM1 and human tissues from DM1 patients tion depended on the presence of MBNL-1 protein. In [104]. Surprisingly, a large 1000-fold discrepancy for live-cell experimental approaches the xtrRNA is likely to transcript number was discovered across mouse models. be over-expressed from an artificial genetic context and In human samples only a few dozen DMPK mRNA mol- may not represent the true dynamics or localization of ecules were detected per cell, with only half of those ex- endogenous repeat expansions. Nonetheless, live and pected to contain the repeat expansion. In a similar fixed cell imaging have revealed that xtrRNA foci are dy- study looking at the abundance and processing of an namic, stable aggregates that likely depend on protein antisense transcript across the DMPK repeat expansion, interactions and may co-localize with known nuclear only a handful of repeat containing antisense transcripts bodies. were quantified per cell [105]. Quantification of the Nuclear bodies can be built around RNA and the mo- repeat-containing intron of C9ORF72 in C9FTD/ALS lecular forces that govern nuclear body formation may patient cells found only a few copies per cell, concluding help explain xtrRNA foci formation and localization. For that each foci might be composed of as few as one example, nuclear paraspeckles depend on the long non- xtrRNA transcript [188]. Therefore, one or a few copies coding RNA NEAT1 (nuclear paraspeckle assembly of xtrRNA may be enough to generate focal aggregates. transcript 1) [321]. Nuclear bodies are essentially Importantly, the stochastic nature of foci formation, membrane-free organelles that are held together by tran- where many cells contain no foci but some contain sev- sient or dynamic protein-protein and protein-RNA inter- eral, suggests that there may be a disproportionate con- actions. These interactions collectively provide a type of tribution to disease for xtrRNA at the individual cell phase separation to organize and compartmentalize cel- level [188]. These reports indicate that knowing the lular processes [336]. It was recently demonstrated that number and type of xtrRNA inside of cells will CAG, CUG and GGGGCC repeat containing RNAs be important for correct interpretation of data and for form soluble aggregates with sol-gel phase separation understanding the role of xtrRNA in disease. properties and behave similar to liquid-like droplets [132]. These properties were dependent on the repeat Nuclear xtrRNA retention and surveillance mechanisms expansion length and base-pairing interactions. In con- The nuclease enzymes primarily responsible for degrad- trast, CCCCGG repeats did not form phase transitions, ing nuclear RNA are the (3'-5' exori- suggesting that not all xtrRNA will possess these proper- bonuclease activity) and 5'-3' 2 (XRN2) ties. Interestingly, guanine-rich nucleic acids are less [67]. These enzymes act as part of a nuclear RNA quality soluble than other nucleic acids and appear to be intrin- control and surveillance pathway that monitors tran- sically aggregate-prone apart from protein, especially scription, splicing, and 3'-end formation of pre-mRNAs, when packing into quartets or higher-order quadruplex as well as their packaging into mRNP particles (Fig. 3) Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 10 of 22

Fig. 3 Possible mechanisms of nuclear and cytoplasmic RNA surveillance, nuclear export, and translation of xtrRNA. RNA containing large repeat expansion sequences may be subject to nuclear RNA surveillance mechanisms, including degradation by the nuclear exosome (1) or the XRN2 5'-3' exoribonuclease (1). Export of xtrRNA likely involves bulk mRNA transport via NXF1 (2b), but may also include alternative mechanisms like CRM1-mediated export (2a) or possibly nuclear envelope budding (2c). Cytoplasmic RNA surveillance mechanisms that may control xtrRNA levels and translation include nonsense-mediated decay (NMD) (3a), no-go decay (NGD) (3b), or nonstop decay (NSD) (3c). Translation of xtrRNA is likely to follow canonical cap-dependent translation (4), especially when repeat expansions are embedded in normal coding regions of an mRNA, but may potentially involve internal entry site (IRES)-like mechanisms (4). RAN translation has been shown to be cap-dependent for some repeat expansions, but complete mechanistic details remain to be determined

[67, 146, 338]. Instead of degradation, these pathways Surveillance mechanisms are also related to transcrip- can also signal for retention of aberrant transcripts in tion or splicing of xtrRNA since these might be expected the nucleus, typically at the site of transcription [52, to trigger degradation [67, 146]. However, the existence 220] but sometimes near nuclear pores [67]. Retention of foci and nuclear export of xtrRNA argue that surveil- at the site of transcription is coupled to nuclear exosome lance mechanisms are incomplete or inefficient for activity, particularly the Rrp6p subunit [66, 220]. The xtrRNA removal. At present it is unknown how many TPR protein, a mammalian ortholog of yeast Mlp1/2p, is molecules of any repeat expansion-containing RNA are implicated in retention at nuclear pores for mRNAs with synthesized versus how many survive to form foci or exit retained introns that normally exit the nucleus through the nucleus for translation. It is likely that repeat the nuclear export factor 1 (NXF1) pathway [49]. Both expansion-containing RNAs survive due to protection of these mechanisms may be relevant to xtrRNA, espe- by protein binding, such as hnRNP proteins [125, 327]. cially when repeat expansions are found in retained Alternatively, factors responsible for recruiting RNA to introns [43, 110, 227]. the nuclear exosome, such as the TRAMP (Trf4/Air2/ Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 11 of 22

Mtr4p ) complex or NEXT (nuclear exo- [82, 302]. NSD could become activated if repeat expan- some targeting) complex, are unable to efficiently sions alter the reading frame or cause loss of the stop recognize and bind the xtrRNA [146, 259]. Thus, codon, such as via mis-splicing. xtrRNA transcripts may escape degradation if they ap- For xtrRNA embedded in mRNAs, either as exons or pear as "normal" mRNPs, having undergone proper RNA introns, mRNA surveillance mechanisms should act to processing like capping, splicing and polyadenylation reduce translation. of these pathways will lead and are associated with appropriate post-processing to degradation by the cytoplasmic version of the exo- factors. some and XRN1, a 5'-3' exoribonuclease. Accessory fac- tors that guide cleavage include the Ski complex Turnover and decay of xtrRNA (composed of Ski2, Ski3, and Ski8 proteins) and the Ski7 An unanswered question remains as to whether foci protein [5, 267]. However, similar to nuclear RNA sur- might contain partially degraded fragments of repeat veillance mechanisms, the cytoplasmic pathways seem RNA in addition to larger intact transcripts. A case in unable to detect or completely remove xtrRNA and pre- point is C9FTD/ALS where the microsatellite expansion vent translation. Methods to enhance RNA surveillance occurs in an intron but nuclear RNA foci and cytoplas- mechanisms might represent reasonable targets for mic translation are both observed. When introns are therapeutic intervention. For example (PTC124) spliced out of pre-mRNA transcripts they are typically is a small molecule drug that increases NMD and might destined for rapid degradation unless they contain a sta- sensitize mRNA surveillance to repeat expansions [73]. bly folding RNA element or recruit RNA binding pro- Some studies have uncovered potential RNA turnover teins [115]. Examples include small nucleolar RNAs and mechanisms associated with repeat expansions. In a re- [56, 203]. It is possible that the structures cent RNAi screen several RNA processing factors were that repeat expansion RNAs form, as well as the proteins identified as suppressors of toxicity in C. elegans ex- that they bind, allow them to persist, accumulate, and pressing a (CUG)123 repeat expansion in a 3'-UTR of aggregate as foci [3]. At present the exact type and num- GFP [88]. These factors included RNases, helicases and ber of xtrRNA species that are trapped in foci versus RNA binding proteins that, when knocked-down, caused free and soluble for any disease is unknown. It is also increased toxicity and enhanced nuclear foci formation. not known whether partially degraded xtrRNA frag- A complex (NPC) protein, npp-4, was also ments are stable enough to accumulate. Furthermore, a suppressor in this screen. Interestingly, smg-2, a con- there is no distinction between what are the major spe- served helicase and central component of the NMD cies or which are nuclear versus cytoplasmic. pathway, was a strong suppressor. Knock-down of NMD When mRNAs that contain microsatellite repeat ex- resulted in a several-fold increase in GFP-(CUG)123 pansions reach the cytoplasm they may encounter add- RNA expression levels and increased GFP translation. itional quality control mechanisms designed to eliminate The substantial increase in 3'-UTR GC-content was aberrant mRNA and prevent its translation. These in- identified as the likely trigger for NMD of the GFP- clude nonsense-mediated decay (NMD), no-go decay (CUG)123 RNA [88]. Smg-2 was also identified as a sup- (NGD), and non-stop decay (NSD) (Fig. 3) [267]. NMD pressor of poly-glutamine aggregation previously, likely recognizes premature stop codons through possibly mul- through NMD of aberrant repeat-containing HTT tran- tiple mechanisms. NMD can be triggered if exon- scripts [328]. These results provide one example of the junction complexes (EJCs), which are deposited near role of RNA turnover in controlling toxicity of xtrRNA splicing junctions, are encountered downstream of a in repeat expansion disorders. [172, 199]. Another mechanism of NMD may involve the relative length of sequence 3' to the stop Nuclear export and translation of xtrRNA codon [4, 172]. Repeat expansions could conceivably Canonical mRNA export pathways alter the positioning of a stop codon or extend the 3' RNA export from the nucleus involves several distinct UTR region and trigger NMD. When ribosome transla- pathways depending on the RNA and the various protein tion is significantly slowed or stalled then NGD can be factors that constitute the RNP particle [273]. For nu- triggered. Stalling is thought to be initiated by unusually clear mRNA there are two main export pathways: stable RNA structures or protein-binding motifs and re- NXF1-mediated and region maintenance 1 sult in endonucleolytic cleavage [63]. Since repeat ex- (CRM1)-mediated, although NXF1 is the primary trans- pansions are believed to fold into stable hairpins or port system for bulk mRNAs (Fig. 3) [60]. Both mecha- tetraplexes they could possibly trigger NGD. In the case nisms rely on adapter proteins to specify the RNA cargo of NSD, the ribosome can become stuck on mRNA that for export. Many of the factors required for successful does not possess a stop codon. These complexes must export are deposited co-transcriptionally and post- be resolved by cleavage and degradation of the mRNA transcriptionally during mRNP assembly [22]. The C- Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 12 of 22

terminal domain of RNA Pol II serves as a docking plat- exports noncoding RNAs like spliceosomal RNA form for a wide variety of mRNA processing and mRNP (snRNA) [10, 74, 131]. There is no reported RNA bind- assembly proteins and plays critical roles in establishing ing affinity of CRM1 so selective export of mRNAs de- mRNP composition [139]. Correct processing of mRNA, pends on the RNA-binding properties of its cargo such as capping, splicing and 3'-end formation, deter- proteins [60]. Export of xtrRNA by CRM1 might only mines the ability of these factors to bind mRNA and as- require that the repeat expansion sequence or structure semble export-competent mRNP particles [36, 60, 335]. somehow recruit a CRM1 cargo protein. The mRNA-associated adapter proteins and com- Export of intronic xtrRNA would be expected to re- plexes represent a complicated matrix of possible in- quire aberrant splicing that resulted in its retention in teractions that dictate export efficiency [22, 60]. NXF1 mRNA, as has been implicated for the intronic C9FTD/ can specifically bind the constitutive transport element ALS repeat expansion [227]. Alternative export pathways found in some mRNAs and viral RNAs, like that of exist but seem unlikely given their very specific nature. the Mason Pfizer monkey virus, to directly facilitate For example, transfer RNA (tRNA) undergoes multiple export [101, 178]. However, for bulk cellular mRNA maturation phases that cumulatively result in two separ- transport NXF1 uses adapters like TREX (transcription ate import and export steps [273]. These export path- export complex) [60, 111]. Although the NXF1 protein ways involve specific RNA-protein interactions, such as interacts loosely with RNA, TREX helps mediate spe- EXP-t and EXP5 [8, 29], that are unlikely to mediate cific binding through its subunit ALY/REF [124], a xtrRNA export. For any export pathway through the protein previously reported to interact with C9ORF72 NPC, xtrRNA must somehow establish RNP complexes GGGGCC RNA repeats [47]. TREX associates with that pass the requisite tests for licensing of export. mRNA during synthesis and processing via mRNA Nuclear exit of RNP granules, such as nuclear xtrRNA capping and splicing events [138, 204] and appears to foci, might also be possible through nuclear envelope be primarily recruited to the 5' ends of mRNAs in hu- budding (Fig. 3). This mechanism involves TorsinA, nu- man cells via interaction between ALY/REF and the clear lamina, and other uncharacterized factors. Nuclear cap binding complex component CBP80 [36]. In budding was discovered as part of the nuclear egress addition to ALY/REF, TREX is composed of the THO mechanism of large nucleocapsid particles of Herpes vi- complex, CIP29, and UAP56, a component of the EJC ruses [55, 78, 200, 221, 277]. Nuclear envelope budding [37, 53, 157]. For repeat expansion disorders, NXF1 has been found to be a natural process for nuclear re- seems to be the most likely pathway since disease- lease of large RNP complexes during development of associated xtrRNA are transcribed from coding gene neuromuscular junctions in Drosophila melanogaster loci and TREX is deposited onto mRNAs early during [135, 277]. However, knock-out of TorsinA in HeLa cells transcription [204]. had little impact on Herpes virus production [294], suggesting alternative factors or mechanisms in hu- Nuclear export of xtrRNA man cells. If xtrRNA is exported by nuclear envelope A recent study connected NXF1 transport of C9FTD/ budding it would have to mimic specific RNP granule ALS intronic xtrRNA via interaction with the export formation that elicits nuclear envelope budding, which adapter SR-rich splicing factor 1 (SRSF1) [110]. SRSF1 at present involves mechanisms that are largely appeared to interact and colocalize with C9ORF72 uncharacterized [78]. xtrRNA. Depletion of SRSF1 prevented neurodegenera- tion in a fly model and suppressed cell death in patient- Translation of xtrRNA derived motor neurons and astrocytes. Depleting SRSF1 If xtrRNA can successfully exit the nucleus it is a po- or preventing interaction with NXF1 inhibited nuclear tential candidate for translation. However, mRNAs export of repeat-containing C9ORF72 transcripts and that contain expanded tandem repeats are possibly blocked RAN translation. Thus, SRSF1 might serve as a the only practical source for translation of repeat ex- therapeutic target in C9FTD/ALS. This report highlights pansion polypeptides since they contain the prerequis- the value of understanding RNA biology in the context ite sequence elements and protein factors to mediate of repeat expansion disorders. canonical cap-dependent translation. These typically Most disease-associated xtrRNA is embedded in ex- include 5' cap structures, bound eukaryotic initiation onic or untranslated regions (Table 1) and therefore factors (eIFs), a poly-A tail, and appropriate mRNP likely exits the nucleus via mRNA export pathways. complexes like the EJC [116, 301, 315]. Translation of CRM1 exports proteins and their associated RNAs via xtrRNA sequence embedded within the coding exon interaction with sequences and of a gene, such as is found in SBMA, HD, DRPLA Ran-GTP (Fig. 3) [60, 74, 77]. CRM1 interacts directly (dentatorubral-pallidoluysian atrophy), OPMD (oculo- with the NPC at the nuclear periphery and commonly pharyngeal muscular dystrophy) and several of the SCA Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 13 of 22

disorders (Table 1), are translated by canonical mecha- translation could even represent a specialized form of nisms. Most repeat expansions form stable secondary uORF translation that is triggered by stable xtrRNA structures that have been shown to reduce the amount of structures. Both mechanisms can initiate at near- overall translation by presumably inducing stalling, cognate start codons (although RAN translation may frame-shifting or abortive translation [72, 222, 244, 313]. use other codons or other mechanisms, like frame- In contrast, the specific binding of MID1 protein to shifting) and are influenced by surrounding sequence huntingtin mRNA, which contains CAG repeat expan- context that might impact RNA folding or protein in- sions, has been reported to enhance translation and teractions [96, 117]. lead to greater levels of aberrant protein [160]. This Recent investigations have demonstrated that certain mechanism has also been proposed to enhance transla- RAN translation products of C9FTD/ALS disrupt the tion of other CAG repeat expansion-containing genes function of membrane-free cellular organelles, such as that cause disease [98]. Canonical translation of repeat stress granules, Cajal bodies and the [174, expansions that are found in-frame in coding se- 182]. These polypeptides seem to block the formation or quences is expected to generate otherwise normal pro- critical interaction dynamics of membrane-free organ- tein that simply contain long tracts of repetitive elles and RNA granules, which are important for neur- polypeptide [296]. onal cell signaling and health [269, 288]. Transport of macromolecules through the nuclear pore complex Repeat-associated non-AUG translation depends on interactions that resemble membrane-free The translation of noncoding xtrRNA irrespective of a organelle structure [207, 219]. They are organized by dy- canonical was recently discovered and namic protein interactions of low complexity domain termed repeat-associated non-AUG (RAN) translation proteins, including phenylalanine-glycine (FG) repeats, [42, 96, 290, 339]. Repeat expansion diseases where this which may explain why certain C9FTD/ALS RAN trans- mechanism has been observed now include SCA2, lation products are reported to disrupt nucleocytoplas- SCA8, SCA31, HD, FXTAS/FXPOI, and C9FTD/ALS mic transport [80, 136, 264, 326]. RAN translation [9, 13, 27, 96, 129, 218, 261, 290, 339, 340]. RAN products can also aggregate and are implicated in the translation of xtrRNA sequence can occur in many disruption of a variety of other pathways [12, 42, 96, contexts, including repeat expansions found in un- 142, 286, 339]. translated regions, retained introns, and even those Several important questions remain concerning the embedded in coding exons [96]. The mechanisms of mechanisms of RAN translation. For example, how simi- RAN translation remain poorly understood and could lar are the mechanisms of RAN translation across di- involve several scenarios, possibly even internal ribosome verse repeat expansion and sequence contexts [42, 96]? entry site (IRES)-like mechanisms (Fig. 3) [96, 339]. For RAN translation maybe a spectrum of related mecha- the CGG repeats of FMR1 that cause FXTAS a more nisms based upon modulation of ribosomal scanning, straightforward mechanism is emerging. In this case, translation initiation, and translation elongation [301]. RAN translation is m7G cap-dependent where a pre- RAN translation can initiate just upstream from the re- initiation complex scans the RNA looking for a start peat expansion, but how often can RAN translation initi- codon [96, 141]. When the CGG repeats are present and ate within the repeat sequence itself [339]? In vitro and stable structures are presumably encountered then stal- cell-based model systems suggest that RAN translation ling occurs and significantly enhances the ability of the can proceed uninterrupted through an entire repeat ex- ribosome to select a near-cognate start codon, or possibly pansion [141, 213, 339, 340]. Yet some expansions are any codon, to initiate translation [141, 154, 158, 339]. A massive in size. Therefore, how often do repeat expan- similar mechanism is favored for the CAG and CUG re- sions induce frame-shifting or possibly even early trans- peats of sense and antisense transcripts in SCA8 [339]. lation termination [313]? Also, what factors are unique This mechanism is proposed to allow translation initi- to RAN translation? Finding answers to these mechanis- ation upstream of a repeat expansion in multiple reading tic questions may be critical for developing future thera- frames [42, 96]. The sequence context, such as the leader peutic molecules that can target and selectively block sequence during scanning, the types of potential near- xtrRNA translation. cognate start codons, and the repeat expansion sequence and size all appear to modulate the degree of RAN trans- Conclusion lation [12, 141, 261, 339]. RNA species that contain simple tandem repeat se- The mechanism of RAN translation may be related to quences occupy an underexplored world of RNA biol- translation of upstream open reading frames (uORFs), ogy. Recent studies have begun to revisit the a widespread phenomena revealed through high- transcription and translation of repeat expansions. How- throughput ribosomal footprint profiling [128]. RAN ever, significant gaps remain for processes like cellular Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 14 of 22

transport and turnover of xtrRNA. Placing repeat expan- 6. Arcot SS, Wang Z, Weber JL, Deininger PL, Batzer MA (1995) Alu repeats: a sion disease mechanism studies in the context of current source for the genesis of primate microsatellites. 29:136–144. doi:10.1006/geno.1995.1224 RNA biology will help reveal a better understanding of 7. Arimbasseri AG, Rijal K, Maraia RJ (2014) Comparative overview of RNA how the cell deals with xtrRNA and identify mechanisms polymerase II and III transcription cycles, with focus on RNA polymerase III unique to repeat expansions. termination and reinitiation. Transcription 5:e27639. doi:10.4161/trns.27369 8. Arts GJ, Kuersten S, Romby P, Ehresmann B, Mattaj IW (1998) The role of Investigations into the biology of xtrRNA promise to exportin-t in selective nuclear export of mature tRNAs. EMBO J 17:7430–7441. unlock new approaches to therapeutics. Transcription doi:10.1093/emboj/17.24.7430 across repeat expansions has opportunities for thera- 9. Ash PE, Bieniek KF, Gendron TF, Caulfield T, Lin WL, Dejesus-Hernandez M, van Blitterswijk MM, Jansen-West K, Paul JW, 3rd, Rademakers Ret al (2013) peutic development, such as modulating the function of Unconventional translation of C9ORF72 GGGGCC expansion generates Supt4h. Likewise, translation of repeat expansions, espe- insoluble polypeptides specific to c9FTD/ALS. Neuron 77: 639-646. cially RAN translation, may become more targetable as doi:10.1016/j.neuron.2013.02.004 10. Bai B, Moore HM, Laiho M (2013) CRM1 and its ribosome export adaptor molecular mechanisms become better characterized and NMD3 localize to the nucleolus and affect rRNA synthesis. Nucleus specific factors identified. Selectively blocking both the 4:315–325. doi:10.4161/nucl.25342 synthesis of xtrRNA or its translation are attractive 11. Bak ST, Sakellariou D, Pena-Diaz J (2014) The dual nature of mismatch repair as antimutator and mutator: for better or for worse. Front Genet 5:287. therapeutic approaches since they could extrapolate to doi:10.3389/fgene.2014.00287 multiple repeat expansion disorders. Turnover of 12. Banez-Coronel M, Ayhan F, Tarabochia AD, Zu T, Perez BA, Tusi SK, xtrRNA should become increasingly important since Pletnikova O, Borchelt DR, Ross CA, Margolis RL et al (2015) RAN Translation – several potential therapeutic strategies employ targeted in Huntington Disease. Neuron 88:667 677. doi:10.1016/j.neuron.2015.10.038 13. Banez-Coronel M, Porta S, Kagerbauer B, Mateu-Huertas E, Pantano L, Ferrer I, and selective degradation of repeat expansion-containing Guzman M, Estivill X, Marti E (2012) A pathogenic mechanism in Huntington's RNA, such as antisense oligonucleotides and small inter- disease involves small CAG-repeated RNAs with neurotoxic activity. PLoS fering RNAs [64, 134, 169, 239, 275]. Genet 8:e1002481. doi:10.1371/journal.pgen.1002481 14. Baralle D, Buratti E (2017) RNA splicing in human disease and in the clinic. Clin Sci (Lond) 131:355–368. doi:10.1042/CS20160211 Funding 15. Baralle M, Pastor T, Bussani E, Pagani F (2008) Influence of Friedreich ataxia This work was supported by a United States Department of Defense GAA noncoding repeat expansions on pre-mRNA processing. Am J Hum Amyotrophic Lateral Sclerosis Research Program (ALSRP) Grant and a Judith Genet 83:77–88. doi:10.1016/j.ajhg.2008.06.018 and Jean Pape Adams Charitable Foundation Grant to K.T.G. 16. Beaudet AL, Meng L (2016) Gene-targeting pharmaceuticals for single-gene disorders. Hum Mol Genet 25:R18–R26. doi:10.1093/hmg/ddv476 Authors’ contributions 17. Bernecky C, Herzog F, Baumeister W, Plitzko JM, Cramer P (2016) Structure – KJR performed an initial literature survey and draft of the manuscript of transcribing mammalian RNA polymerase II. Nature 529:551 554. and prepared tables and figures. KTG performed an in-depth literature doi:10.1038/nature16482 survey, finalized tables and figures, and wrote the final draft of the 18. Biancalana V, Taine L, Bouix JC, Finck S, Chauvin A, De Verneuil H, Knight SJ, manuscript. Both authors read and approved the final manuscript. Stoll C, Lacombe D, Mandel JL (1996) Expansion and methylation status at FRAXE can be detected on EcoRI blots used for FRAXA diagnosis: analysis of four FRAXE families with mild mental retardation in males. Am J Hum Competing interests Genet 59:847–854 The authors declare that they have no competing interests. 19. Bidichandani SI, Ashizawa T, Patel PI (1998) The GAA triplet-repeat expansion in Friedreich ataxia interferes with transcription and may be associated with an unusual DNA structure. Am J Hum Genet 62:111–121. Publisher’sNote doi:10.1086/301680 Springer Nature remains neutral with regard to jurisdictional claims in 20. Biffi G, Di Antonio M, Tannahill D, Balasubramanian S (2014) Visualization published maps and institutional affiliations. and selective chemical targeting of RNA G-quadruplex structures in the cytoplasm of human cells. Nat Chem 6:75–80. doi:10.1038/nchem.1805 Received: 28 June 2017 Accepted: 22 August 2017 21. Biyani M, Nishigaki K (2005) Structural characterization of ultra-stable higher- ordered aggregates generated by novel guanine-rich DNA sequences. Gene 364:130–138. doi:10.1016/j.gene.2005.05.041 References 22. Bjork P, Wieslander L (2014) Mechanisms of mRNA export. Semin Cell Dev 1. Abecasis GR, Auton A, Brooks LD, DePristo MA, Durbin RM, Handsaker RE, Biol 32:47–54. doi:10.1016/j.semcdb.2014.04.027 Kang HM, Marth GT, McVean GA (2012) An integrated map of genetic 23. Brais B, Bouchard JP, Xie YG, Rochefort DL, Chretien N, Tome FM, variation from 1,092 human . Nature 491:56–65. doi:10.1038/ Lafreniere RG, Rommens JM, Uyama E, Nohira Oet al (1998) Short GCG nature11632 expansions in the PABP2 gene cause oculopharyngeal muscular 2. Adihe Lokanga R, Zhao XN, Entezam A, Usdin K (2014) X inactivation plays a dystrophy. Nat Genet 18:164–167. doi:10.1038/ng0298-164 major role in the gender bias in somatic expansion in a mouse model of 24. Brinkmann B, Klintschar M, Neuhuber F, Huhne J, Rolf B (1998) the fragile X-related disorders: implications for the mechanism of repeat in human microsatellites: influence of the structure and length of the expansion. Hum Mol Genet 23:4985–4994. doi:10.1093/hmg/ddu213 tandem repeat. Am J Hum Genet 62:1408–1415. doi:10.1086/301869 3. Akiyama BM, Eiler D, Kieft JS (2016) Structured RNAs that evade or 25. Brouwer JR, Huguet A, Nicole A, Munnich A, Gourdon G (2013) confound exonucleases: function follows form. Curr Opin Struct Biol Transcriptionally Repressive Chromatin Remodelling and CpG 36:40–47. doi:10.1016/j.sbi.2015.12.006 Methylation in the Presence of Expanded CTG-Repeats at the DM1 4. Amrani N, Ganesan R, Kervestin S, Mangus DA, Ghosh S, Jacobson A (2004) Locus. J Nucleic Acids 2013:567435. doi:10.1155/2013/567435 A faux 3'-UTR promotes aberrant termination and triggers nonsense- 26. Budworth H, McMurray CT (2013) Bidirectional transcription of trinucleotide mediated mRNA decay. Nature 432:112–118. doi:10.1038/nature03060 repeats: roles for excision repair. DNA repair 12:672–684. doi:10.1016/j. 5. Anderson JS, Parker RP (1998) The 3' to 5' degradation of yeast mRNAs is a dnarep.2013.04.019 general mechanism for mRNA turnover that requires the SKI2 DEVH box 27. Buijsen RA, Visser JA, Kramer P, Severijnen EA, Gearing M, Charlet- protein and 3' to 5' exonucleases of the exosome complex. EMBO J Berguerand N, Sherman SL, Berman RF, Willemsen R, Hukema RK (2016) 17:1497–1506. doi:10.1093/emboj/17.5.1497 Presence of inclusions positive for polyglycine containing protein, Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 15 of 22

FMRpolyG, indicates that repeat-associated non-AUG translation plays a role Antisense RNA foci in the motor neurons of C9ORF72-ALS patients are in fragile X-associated primary ovarian insufficiency. Hum Reprod 31:158–168. associated with TDP-43 proteinopathy. Acta Neuropathol 130:63–75. doi:10.1093/humrep/dev280 doi:10.1007/s00401-015-1429-9 28. Buschiazzo E, Gemmell NJ (2006) The rise, fall and renaissance of microsatellites 47. Cooper-Knock J, Walsh MJ, Higginbottom A, Robin Highley J, Dickman MJ, in eukaryotic genomes. BioEssays : news and reviews in molecular, cellular and Edbauer D, Ince PG, Wharton SB, Wilson SA, Kirby J et al (2014) developmental biology 28:1040–1050. doi:10.1002/bies.20470 Sequestration of multiple RNA recognition motif-containing proteins by 29. Calado A, Treichel N, Muller EC, Otto A, Kutay U (2002) Exportin-5-mediated C9orf72 repeat expansions. Brain 137:2040-2051. doi:10.1093/brain/awu120 nuclear export of eukaryotic 1A and tRNA. EMBO J 48. Cortes CJ, La Spada AR (2015) Autophagy in polyglutamine disease: 21:6216–6224 Imposing order on disorder or contributing to the chaos? Mol Cell Neurosci 30. Campuzano V, Montermini L, Molto MD, Pianese L, Cossee M, Cavalcanti F, 66:53–61. doi:10.1016/j.mcn.2015.03.010 Monros E, Rodius F, Duclos F, Monticelli A et al (1996) Friedreich's ataxia: 49. Coyle JH, Bor YC, Rekosh D, Hammarskjold ML (2011) The Tpr protein autosomal recessive disease caused by an intronic GAA triplet repeat regulates export of mRNAs with retained introns that traffic through the expansion. Science 271:1423–1427 Nxf1 pathway. RNA 17:1344–1356. doi:10.1261/.2616111 31. Chaisson MJ, Huddleston J, Dennis MY, Sudmant PH, Malig M, Hormozdiari F, 50. Crickard JB, Fu J, Reese JC (2016) Biochemical Analysis of Yeast Suppressor Antonacci F, Surti U, Sandstrom R, Boitano M et al (2015) Resolving the of Ty 4/5 (Spt4/5) Reveals the Importance of Nucleic Acid Interactions in the complexity of the human genome using single-molecule sequencing. Nature Prevention of RNA Polymerase II Arrest. J Biol Chem 291:9853–9870. 517:608–611. doi:10.1038/nature13907 doi:10.1074/jbc.M116.716001 32. Chakraborty R, Kimmel M, Stivers DN, Davison LJ, Deka R (1997) Relative 51. Crouau-Roy B, Clisson I (2000) Evolution of an Alu DNA element of type Sx mutation rates at di-, tri-, and tetranucleotide microsatellite loci. Proc Natl in the lineage of primates and the origin of an associated tetranucleotide Acad Sci U S A 94:1041–1046 microsatellite. Genome 43:642–648 33. Chan NL, Guo J, Zhang T, Mao G, Hou C, Yuan F, Huang J, Zhang Y, Wu J, 52. Custodio N, Carmo-Fonseca M, Geraghty F, Pereira HS, Grosveld F, Gu L et al (2013) Coordinated processing of 3' slipped (CAG)n/(CTG)n Antoniou M (1999) Inefficient processing impairs release of RNA from the site hairpins by DNA polymerases beta and delta preferentially induces repeat of transcription. EMBO J 18:2855–2866. doi:10.1093/emboj/18.10.2855 expansions. J Biol Chem 288:15015–15022. doi:10.1074/jbc.M113.464370 53. Custodio N, Carvalho C, Condado I, Antoniou M, Blencowe BJ, Carmo- 34. Charlet BN, Savkur RS, Singh G, Philips AV, Grice EA, Cooper TA (2002) Loss Fonseca M (2004) In vivo recruitment of exon junction complex proteins to of the muscle-specific chloride channel in type 1 myotonic dystrophy due transcription sites in mammalian cell nuclei. RNA 10:622–633 to misregulated alternative splicing. Mol Cell 10:45–53 54. Dai WJ, Zhu LY, Yan ZY, Xu Y, Wang QL, Lu XJ (2016) CRISPR-Cas9 for in 35. Chen IC, Lin HY, Lee GC, Kao SH, Chen CM, Wu YR, Hsieh-Li HM, Su MT, vivo Gene Therapy: Promise and Hurdles. Mol Ther Nucleic Acids Lee-Chen GJ (2009) Spinocerebellar ataxia type 8 larger triplet expansion 5:e349. doi:10.1038/mtna.2016.58 alters histone modification and induces RNA foci. BMC Mol Biol 10:9. 55. Darlington RW, Moss LH 3rd (1968) Herpesvirus envelopment. J Virol 2:48–55 doi:10.1186/1471-2199-10-9 56. Daugaard I, Hansen TB (2017) Biogenesis and Function of Ago-Associated 36. Cheng H, Dufu K, Lee CS, Hsu JL, Dias A, Reed R (2006) Human mRNA RNAs. Trends Genet. doi:10.1016/j.tig.2017.01.003 export machinery recruited to the 5' end of mRNA. Cell 127:1389–1400. 57. David G, Abbas N, Stevanin G, Durr A, Yvert G, Cancel G, Weber C, Imbert G, doi:10.1016/j.cell.2006.10.044 Saudou F, Antoniou Eet al (1997) of the SCA7 gene reveals a highly 37. Chi B, Wang Q, Wu G, Tan M, Wang L, Shi M, Chang X, Cheng H (2013) Aly unstable CAG repeat expansion. Nat Genet 17:65–70. doi:10.1038/ng0997-65 and THO are required for assembly of the human TREX complex and 58. de la Mata M, Alonso CR, Kadener S, Fededa JP, Blaustein M, Pelisch F, association of TREX components with the spliced mRNA. Nucleic Acids Res Cramer P, Bentley D, Kornblihtt AR (2003) A slow RNA polymerase II affects 41:1294–1306. doi:10.1093/nar/gks1188 alternative splicing in vivo. Mol Cell 12:525–532 38. Childs-Disney JL, Hoskins J, Rzuczek SG, Thornton CA, Disney MD (2012) 59. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rationally designed small molecules targeting the RNA that causes Rutherford NJ, Nicholson AM, Finch NA, Flynn H, Adamson J et al (2011) myotonic dystrophy type 1 are potently bioactive. Acs Chem Biol 7:856–862. Expanded GGGGCC Hexanucleotide Repeat in Noncoding Region of doi:10.1021/cb200408a C9ORF72 Causes Chromosome 9p-Linked FTD and ALS. Neuron 72:245–256. 39. Childs-Disney JL, Yildirim I, Park H, Lohman JR, Guan L, Tran T, Sarkar P, doi:10.1016/j.neuron.2011.09.011 Schatz GC, Disney MD (2014) Structure of the myotonic dystrophy type 2 60. Delaleau M, Borden KL (2015) Multiple Export Mechanisms for mRNAs. Cells RNA and designed small molecules that reduce toxicity. Acs Chem Biol 4:452–473. doi:10.3390/cells4030452 9:538–550. doi:10.1021/cb4007387 61. Diegoli TM (2015) Forensic typing of short tandem repeat markers on the X 40. Chung DW, Rudnicki DD, Yu L, Margolis RL (2011) A natural antisense and Y . Forensic Sci Int Genet 18:140–151. doi:10.1016/j.fsigen. transcript at the Huntington's disease repeat locus regulates HTT expression. 2015.03.013 Hum Mol Genet 20:3467–3477. doi:10.1093/hmg/ddr263 62. Dodd DW, Tomchick DR, Corey DR, Gagnon KT (2016) Pathogenic C9ORF72 41. Clark RM, Dalgliesh GL, Endres D, Gomez M, Taylor J, Bidichandani SI (2004) Antisense Repeat RNA Forms a Double Helix with Tandem C:C Mismatches. Expansion of GAA triplet repeats in the human genome: unique origin of Biochemistry 55:1283–1286. doi:10.1021/acs.biochem.6b00136 the FRDA mutation at the center of an Alu. Genomics 83:373–383. 63. Doma MK, Parker R (2006) Endonucleolytic cleavage of eukaryotic mRNAs doi:10.1016/j.ygeno.2003.09.001 with stalls in translation elongation. Nature 440:561–564. doi:10.1038/ 42. Cleary JD, Ranum LP (2014) Repeat associated non-ATG (RAN) translation: nature04530 new starts in microsatellite expansion disorders. Curr Opin Genet Dev 64. Donnelly CJ, Zhang PW, Pham JT, Haeusler AR, Mistry NA, Vidensky S, 26C:6–15. doi:10.1016/j.gde.2014.03.002 Daley EL, Poth EM, Hoover B, Fines DM et al (2013) RNA toxicity from 43. Cohen-Hadad Y, Altarescu G, Eldar-Geva T, Levi-Lahad E, Zhang M, Rogaeva E, the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Gotkine M, Bartok O, Ashwal-Fluss R, Kadener S et al (2016) Marked Differences Neuron 80:415–428. doi:10.1016/j.neuron.2013.10.015 in C9orf72 Methylation Status and Isoform Expression between C9/ALS Human 65. Dumache R, Ciocan V, Muresan C, Enache A (2016) Molecular DNA Analysis Embryonic and Induced Pluripotent Stem Cells. Stem Cell Rep 7:927–940. in . Clin Lab 62:245–248 doi:10.1016/j.stemcr.2016.09.011 66. Eberle AB, Hessle V, Helbig R, Dantoft W, Gimber N, Visa N (2010) Splice-site 44. Colak D, Zaninovic N, Cohen MS, Rosenwaks Z, Yang WY, Gerhardt J, mutations cause Rrp6-mediated nuclear retention of the unspliced RNAs Disney MD, Jaffrey SR (2014) Promoter-bound trinucleotide repeat and transcriptional down-regulation of the splicing-defective genes. PLoS mRNA drives epigenetic silencing in fragile X syndrome. Science One 5:e11540. doi:10.1371/journal.pone.0011540 343:1002–1005. doi:10.1126/science.1245831 67. Eberle AB, Visa N (2014) Quality control of mRNP biogenesis: networking at 45. Conlon EG, Lu L, Sharma A, Yamazaki T, Tang T, Shneider NA, Manley JL the transcription site. Semin Cell Dev Biol 32:37–46. doi:10.1016/j.semcdb. (2016) The C9ORF72 GGGGCC expansion forms RNA G-quadruplex 2014.03.033 inclusions and sequesters hnRNP H to disrupt splicing in ALS brains. eLife 5. 68. Ellegren H (2000) Heterogeneous mutation processes in human microsatellite doi:10.7554/eLife.17820 DNA sequences. Nat Genet 24:400–402. doi:10.1038/74249 46. Cooper-Knock J, Higginbottom A, Stopford MJ, Highley JR, Ince PG, 69. Ellegren H (2004) Microsatellites: simple sequences with complex evolution. Wharton SB, Pickering-Brown S, Kirby J, Hautbergue GM, Shaw PJ (2015) Nat Rev Genet 5:435–445. doi:10.1038/nrg1348 Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 16 of 22

70. Evans-Galea MV, Hannan AJ, Carrodus N, Delatycki MB, Saffery R (2013) 93. Gordenin DA, Kunkel TA, Resnick MA (1997) Repeat expansion–all in a flap? Epigenetic modifications in trinucleotide repeat diseases. Trends Mol Med Nat Genet 16:116–118. doi:10.1038/ng0697-116 19:655–663. doi:10.1016/j.molmed.2013.07.007 94. Grabczyk E, Usdin K (2000) The GAA*TTC triplet repeat expanded in 71. Fellmann C, Gowen BG, Lin PC, Doudna JA, Corn JE (2017) Cornerstones of Friedreich's ataxia impedes transcription elongation by T7 RNA CRISPR-Cas in drug discovery and therapy. Nat Rev Drug Discov 16:89–100. polymerase in a length and supercoil dependent manner. Nucleic Acids doi:10.1038/nrd.2016.238 Res 28:2815–2822 72. Feng Y, Zhang F, Lokey LK, Chastain JL, Lakkis L, Eberhart D, Warren ST 95. Grandi FC, An W (2013) Non-LTR and microsatellites: (1995) Translational suppression by trinucleotide repeat expansion at FMR1. Partners in genomic variation. Mobile Genet Elem 3:e25674. doi:10.4161/ Science 268:731–734 mge.25674 73. Finkel RS (2010) Read-through strategies for suppression of nonsense 96. Green KM, Linsalata AE, Todd PK (2016) RAN translation-What makes it run? mutations in Duchenne/ Becker muscular dystrophy: Brain Res. doi:10.1016/j.brainres.2016.04.003 and ataluren (PTC124). J Child Neurol 25:1158–1164. doi:10.1177/ 97. Greene E, Mahishi L, Entezam A, Kumari D, Usdin K (2007) Repeat-induced 0883073810371129 epigenetic changes in intron 1 of the frataxin gene and its consequences in 74. Floer M, Blobel G (1999) Putative reaction intermediates in Crm1-mediated Friedreich ataxia. Nucleic Acids Res 35:3383–3390. doi:10.1093/nar/gkm271 nuclear protein export. J Biol Chem 274:16279–16286 98. Griesche N, Schilling J, Weber S, Rohm M, Pesch V, Matthes F, Auburger G, 75. Foiry L, Dong L, Savouret C, Hubert L, te Riele H, Junien C, Gourdon G Krauss S (2016) Regulation of mRNA Translation by MID1: A Common (2006) Msh3 is a limiting factor in the formation of intergenerational CTG Mechanism of Expanded CAG Repeat RNAs. Front Cell Neurosci 10:226. expansions in DM1 transgenic mice. Hum Genet 119:520–526. doi:10.1007/ doi:10.3389/fncel.2016.00226 s00439-006-0164-7 99. Groh M, Lufino MM, Wade-Martins R, Gromak N (2014) R-loops associated 76. Follonier C, Oehler J, Herrador R, Lopes M (2013) Friedreich's ataxia- with triplet repeat expansions promote in Friedreich ataxia associated GAA repeats induce replication-fork reversal and unusual and fragile X syndrome. PLoS Genet 10:e1004318. doi:10.1371/journal.pgen. molecular junctions. Nat Struct Mol Biol 20:486–494. doi:10.1038/nsmb.2520 1004318 77. Fornerod M, Ohno M, Yoshida M, Mattaj IW (1997) CRM1 is an export 100. Groh M, Silva LM, Gromak N (2014) Mechanisms of transcriptional receptor for leucine-rich nuclear export signals. Cell 90:1051–1060 dysregulation in repeat expansion disorders. Biochem Soc Trans 78. Fradkin LG, Budnik V (2016) This bud's for you: mechanisms of cellular 42:1123–1128. doi:10.1042/BST20140049 nucleocytoplasmic trafficking via nuclear envelope budding. Curr Opin Cell 101. Gruter P, Tabernero C, von Kobbe C, Schmitt C, Saavedra C, Bachi A, Wilm M, Biol 41:125–131. doi:10.1016/j.ceb.2016.05.001 Felber BK, Izaurralde E (1998) TAP, the human homolog of Mex67p, mediates 79. Fratta P, Mizielinska S, Nicoll AJ, Zloh M, Fisher EMC, Parkinson G, Isaacs AM CTE-dependent RNA export from the nucleus. Mol Cell 1:649–659 (2012) C9orf72 hexanucleotide repeat associated with amyotrophic lateral 102. Guan L, Disney MD (2012) Recent advances in developing small molecules sclerosis and frontotemporal dementia forms RNA G-quadruplexes. Sci Rep- targeting RNA. Acs Chem Biol 7:73–86. doi:10.1021/cb200447r Uk 2:1016. doi:10.1038/Srep01016 103. Gudanis D, Popenda L, Szpotkowski K, Kierzek R, Gdaniec Z (2016) Structural 80. Freibaum BD, Lu Y, Lopez-Gonzalez R, Kim NC, Almeida S, Lee KH, characterization of a dimer of RNA duplexes composed of 8-bromoguanosine Badders N, Valentine M, Miller BL, Wong PC et al (2015) GGGGCC modified CGG trinucleotide repeats: a novel architecture of RNA quadruplexes. repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nucleic Acids Res 44:2409–2416. doi:10.1093/nar/gkv1534 Nature 525:129–133. doi:10.1038/nature14974 104. Gudde AE, Gonzalez-Barriga A, van den Broek WJ, Wieringa B, Wansink DG 81. Freudenreich CH, Kantrow SM, Zakian VA (1998) Expansion and length- (2016) A low absolute number of expanded transcripts is involved in dependent fragility of CTG repeats in yeast. Science 279:853–856 myotonic dystrophy type 1 manifestation in muscle. Hum Mol Genet 82. Frischmeyer PA, van Hoof A, O'Donnell K, Guerrerio AL, Parker R, Dietz HC 25:1648–1662. doi:10.1093/hmg/ddw042 (2002) An mRNA surveillance mechanism that eliminates transcripts lacking 105. Gudde AE, van Heeringen SJ, de Oude AI, van Kessel ID, Estabrook J, termination codons. Science 295:2258–2261. doi:10.1126/science.1067338 Wang ET, Wieringa B, Wansink DG (2017) Antisense transcription of the 83. Froehlich AC, Liu Y, Loros JJ, Dunlap JC (2002) White Collar-1, a circadian myotonic dystrophy locus yields low-abundant RNAs with and without blue light photoreceptor, binding to the promoter. Science (CAG)n repeat. RNA Biol 0. doi:10.1080/15476286.2017.1279787 297:815–819. doi:10.1126/science.1073681 106. Guo J, Gu L, Leffak M, Li GM (2016) MutSbeta promotes trinucleotide repeat 84. Gadgil R, Barthelemy J, Lewis T, Leffak M (2017) Replication stalling and expansion by recruiting DNA polymerase beta to nascent (CAG)n or (CTG)n DNA microsatellite instability. Biophys Chem 225:38–48. doi:10.1016/j.bpc. hairpins for error-prone DNA synthesis. Cell Res 26:775–786. doi:10.1038/cr. 2016.11.007 2016.66 85. Gagnon KT, Corey DR (2015) Stepping toward therapeutic CRISPR. Proc Natl 107. Guo JU, Bartel DP (2016) RNA G-quadruplexes are globally unfolded in Acad Sci U S A 112:15536–15537. doi:10.1073/pnas.1521670112 eukaryotic cells and depleted in bacteria. Science 353. doi:10.1126/science. 86. Gan W, Guan Z, Liu J, Gui T, Shen K, Manley JL, Li X (2011) R-loop-mediated aaf5371 genomic instability is caused by impairment of replication fork progression. 108. Haeusler AR, Donnelly CJ, Periz G, Simko EA, Shaw PG, Kim MS, Maragakis NJ, Genes Dev 25:2041–2056. doi:10.1101/gad.17010011 Troncoso JC, Pandey A, Sattler Ret al (2014) C9orf72 nucleotide repeat 87. Garavis M, Gonzalez C, Villasante A (2013) On the origin of the eukaryotic structures initiate molecular cascades of disease. Nature 507:195–200. chromosome: the role of noncanonical DNA structures in doi:10.1038/nature13124 evolution. Genome Biol Evol 5:1142–1150. doi:10.1093/gbe/evt079 109. Hamperl S, Cimprich KA (2016) Conflict Resolution in the Genome: How 88. Garcia SM, Tabach Y, Lourenco GF, Armakola M, Ruvkun G (2014) Identification Transcription and Replication Make It Work. Cell 167:1455–1467. doi:10.1016/ of genes in toxicity pathways of trinucleotide-repeat RNA in C. elegans. Nat j.cell.2016.09.053 Struct Mol Biol 21:712–720. doi:10.1038/nsmb.2858 110. Hautbergue GM, Castelli LM, Ferraiuolo L, Sanchez-Martinez A, Cooper- 89. Gellert M, Lipsett MN, Davies DR (1962) Helix formation by guanylic acid. Knock J, Higginbottom A, Lin YH, Bauer CS, Dodd JE, Myszczynska MA et al Proc Natl Acad Sci U S A 48:2013–2018 (2017) SRSF1-dependent nuclear export inhibition of C9ORF72 repeat 90. Gendron TF, Bieniek KF, Zhang YJ, Jansen-West K, Ash PE, Caulfield T, transcripts prevents neurodegeneration and associated motor deficits. Nat Daughrity L, Dunmore JH, Castanedes-Casey M, Chew J et al (2013) Commun 8:16063. doi:10.1038/ncomms16063 Antisense transcripts of the expanded C9ORF72 hexanucleotide repeat 111. Hautbergue GM, Hung ML, Golovanov AP, Lian LY, Wilson SA (2008) Mutually form nuclear RNA foci and undergo repeat-associated non-ATG exclusive interactions drive handover of mRNA from export adaptors to TAP. translation in c9FTD/ALS. Acta Neuropathol 126:829–844. doi:10.1007/ Proc Natl Acad Sci U S A 105:5154–5159. doi:10.1073/pnas.0709167105 s00401-013-1192-8 112. He F, Todd PK (2011) in nucleotide repeat expansion disorders. 91. Ginno PA, Lott PL, Christensen HC, Korf I, Chedin F (2012) R-loop formation Semin Neurol 31:470–483. doi:10.1055/s-0031-1299786 is a distinctive characteristic of unmethylated human CpG island promoters. 113. He Y, Vogelstein B, Velculescu VE, Papadopoulos N, Kinzler KW (2008) Mol Cell 45:814–825. doi:10.1016/j.molcel.2012.01.017 The antisense of human cells. Science 322:1855–1857. 92. Goers ES, Purcell J, Voelker RB, Gates DP, Berglund JA (2010) MBNL1 binds doi:10.1126/science.1163853 GC motifs embedded in to regulate alternative splicing. Nucleic 114. Heitz D, Rousseau F, Devys D, Saccone S, Abderrahim H, Le Paslier D, Acids Res 38:2467–2484. doi:10.1093/nar/gkp1209 Cohen D, Vincent A, Toniolo D, Della Valle G et al (1991) Isolation of Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 17 of 22

sequences that span the fragile X and identification of a fragile X- ribonucleoprotein granules at the nuclear envelope. Cell Rep 3:988–995. related CpG island. Science 251:1236–1239 doi:10.1016/j.celrep.2013.03.015 115. Hesselberth JR (2013) that introns lead after splicing. Wiley Interdiscip 136. Jovicic A, Mertens J, Boeynaems S, Bogaert E, Chai N, Yamada SB, Paul JW, Rev RNA 4:677–691. doi:10.1002/wrna.1187 3rd, Sun S, Herdy JR, Bieri Get al (2015) Modifiers of C9orf72 dipeptide 116. Hinnebusch AG (2014) The scanning mechanism of repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. initiation. Annu Rev Biochem 83:779–812. doi:10.1146/annurev-biochem- Nat Neurosci 18: 1226-1229 Doi doi:10.1038/nn.4085 060713-035802 137. Kang S, Jaworski A, Ohshima K, Wells RD (1995) Expansion and of 117. Hinnebusch AG, Ivanov IP, Sonenberg N (2016) Translational control by 5'- CTG repeats from human disease genes are determined by the direction of untranslated regions of eukaryotic mRNAs. Science 352:1413–1416. replication in E. coli. Nat Genet 10:213–218. doi:10.1038/ng0695-213 doi:10.1126/science.aad9868 138. Katahira J (2012) mRNA export and the TREX complex. Biochimica Et 118. Hino S, Kondo S, Sekiya H, Saito A, Kanemoto S, Murakami T, Chihara K, Biophysica Acta 1819:507–513. doi:10.1016/j.bbagrm.2011.12.001 Aoki Y, Nakamori M, Takahashi MP et al (2007) Molecular mechanisms 139. Katahira J (2015) Nuclear export of messenger RNA. Genes 6:163–184. responsible for aberrant splicing of SERCA1 in myotonic dystrophy type doi:10.3390/genes6020163 1. Hum Mol Genet 16:2834–2843 doi:10.1093/hmg/ddm239 140. Kawaguchi Y, Okamoto T, Taniwaki M, Aizawa M, Inoue M, Katayama S, 119. Hirtreiter A, Damsma GE, Cheung AC, Klose D, Grohmann D, Vojnic E, Kawakami H, Nakamura S, Nishimura M, Akiguchi I et al (1994) CAG Martin AC, Cramer P, Werner F (2010) Spt4/5 stimulates transcription expansions in a novel gene for Machado-Joseph disease at chromosome elongation through the RNA polymerase clamp coiled-coil motif. 14q32.1 Nat Genet 8:221–228. doi:10.1038/ng1194-221 Nucleic Acids Res 38:4040–4051. doi:10.1093/nar/gkq135 141. Kearse MG, Green KM, Krans A, Rodriguez CM, Linsalata AE, Goldstrohm AC, 120. Holmes SE, O'Hearn E, Rosenblatt A, Callahan C, Hwang HS, Ingersoll- Todd PK (2016) CGG Repeat-Associated Non-AUG Translation Utilizes a Cap- Ashworth RG, Fleisher A, Stevanin G, Brice A, Potter NT et al (2001) A repeat Dependent Scanning Mechanism of Initiation to Produce Toxic Proteins. expansion in the gene encoding junctophilin-3 is associated with Mol Cell 62:314–322. doi:10.1016/j.molcel.2016.02.034 Huntington disease-like 2. Nat Genet 29:377–378. doi:10.1038/ng760 142. Kearse MG, Todd PK (2014) Repeat-associated non-AUG translation and its 121. Holmes SE, O'Hearn EE, McInnis MG, Gorelick-Feldman DA, Kleiderlein JJ, impact in neurodegenerative disease. Neurotherapeutics 11:721–731. Callahan C, Kwak NG, Ingersoll-Ashworth RG, Sherr M, Sumner AJ et al doi:10.1007/s13311-014-0292-z (1999) Expansion of a novel CAG trinucleotide repeat in the 5' region of 143. Kenneson A, Zhang F, Hagedorn CH, Warren ST (2001) Reduced FMRP and PPP2R2B is associated with SCA12. Nat Genet 23:391–392. doi:10.1038/ increased FMR1 transcription is proportionally associated with CGG repeat 70493 number in intermediate-length and premutation carriers. Hum Mol Genet 122. Houseley J, Tollervey D (2011) Repeat expansion in the budding yeast 10:1449–1454 ribosomal DNA can occur independently of the canonical homologous 144. Kerrest A, Anand RP, Sundararajan R, Bermejo R, Liberi G, Dujon B, recombination machinery. Nucleic Acids Res 39:8778–8791. doi:10.1093/nar/ Freudenreich CH, Richard GF (2009) SRS2 and SGS1 prevent chromosomal gkr589 breaks and stabilize triplet repeats by restraining recombination. Nat Struct 123. Huertas P, Aguilera A (2003) Cotranscriptionally formed DNA:RNA hybrids Mol Biol 16:159–167. doi:10.1038/nsmb.1544 mediate transcription elongation impairment and transcription-associated 145. Khorkova O, Myers AJ, Hsiao J, Wahlestedt C (2014) Natural antisense recombination. Mol Cell 12:711–721 transcripts. Hum Mol Genet 23:R54–R63. doi:10.1093/hmg/ddu207 124. Hung ML, Hautbergue GM, Snijders AP, Dickman MJ, Wilson SA (2010) 146. Kilchert C, Wittmann S, Vasiljeva L (2016) The regulation and functions of Arginine methylation of REF/ALY promotes efficient handover of mRNA to the nuclear RNA exosome complex. Nat Rev Mol Cell Biol 17:227–239. TAP/NXF1. Nucleic Acids Res 38:3351–3361. doi:10.1093/nar/gkq033 doi:10.1038/nrm.2015.15 125. Iglesias N, Stutz F (2008) Regulation of mRNP dynamics along the export 147. Kiliszek A, Rypniewski W (2014) Structural studies of CNG repeats. Nucleic pathway. FEBS letters 582:1987–1996. doi:10.1016/j.febslet.2008.03.038 Acids Res 42:8189–8199 126. Ikeda Y, Daughters RS, Ranum LP (2008) Bidirectional expression of the 148. Kim JC, Harris ST, Dinter T, Shah KA, Mirkin SM (2017) The role of break- SCA8 expansion mutation: one mutation, two genes. Cerebellum 7:150–158. induced replication in large-scale expansions of (CAG)n/(CTG)n repeats. Nat doi:10.1007/s12311-008-0010-7 Struct Mol Biol 24:55–60. doi:10.1038/nsmb.3334 127. Imbert G, Saudou F, Yvert G, Devys D, Trottier Y, Garnier JM, Weber C, 149. Kim JC, Mirkin SM (2013) The balancing act of DNA repeat expansions. Curr Mandel JL, Cancel G, Abbas N et al (1996) Cloning of the gene for Opin Genet Dev 23:280–288. doi:10.1016/j.gde.2013.04.009 spinocerebellar ataxia 2 reveals a locus with high sensitivity to expanded 150. Kino Y, Washizu C, Kurosawa M, Oma Y, Hattori N, Ishiura S, Nukina N (2015) CAG/glutamine repeats. Nat Genet 14:285–291. doi:10.1038/ng1196-285 Nuclear localization of MBNL1: splicing-mediated autoregulation and 128. Ingolia NT, Ghaemmaghami S, Newman JR, Weissman JS (2009) Genome- repression of repeat-derived aberrant proteins. Hum Mol Genet 24:740–756. wide analysis in vivo of translation with nucleotide resolution using doi:10.1093/hmg/ddu492 ribosome profiling. Science 324:218–223. doi:10.1126/science.1168978 151. Klein BJ, Bose D, Baker KJ, Yusoff ZM, Zhang X, Murakami KS (2011) RNA 129. Ishiguro T, Sato N, Ueyama M, Fujikake N, Sellier C, Kanegami A, Tokuda E, polymerase and transcription elongation factor Spt4/5 complex structure. Zamiri B, Gall-Duncan T, Mirceta M et al (2017) Regulatory Role of RNA Proc Natl Acad Sci U S A 108:546–550. doi:10.1073/pnas.1013828108 Chaperone TDP-43 for RNA Misfolding and Repeat-Associated Translation in 152. Knight SJ, Flannery AV, Hirst MC, Campbell L, Christodoulou Z, Phelps SR, SCA31. Neuron. doi:10.1016/j.neuron.2017.02.046 Pointon J, Middleton-Price HR, Barnicoat A, Pembrey ME et al (1993) 130. Iyer RR, Pluciennik A, Napierala M, Wells RD (2015) DNA Triplet Repeat Trinucleotide repeat amplification and hypermethylation of a CpG island in Expansion and Mismatch Repair. Annu Rev Biochem. doi:10.1146/annurev- FRAXE mental retardation. Cell 74:127–134 biochem-060614-034010 153. Kobayashi H, Abe K, Matsuura T, Ikeda Y, Hitomi T, Akechi Y, Habu T, Liu W, 131. Izumi H, McCloskey A, Shinmyozu K, Ohno M (2014) p54nrb/NonO and PSF Okuda H, Koizumi A (2011) Expansion of intronic GGCCTG hexanucleotide promote U snRNA nuclear export by accelerating its export complex repeat in NOP56 causes SCA36, a type of spinocerebellar ataxia accompanied assembly. Nucleic Acids Res 42:3998–4007. doi:10.1093/nar/gkt1365 by motor neuron involvement. Am J Hum Genet 89:121–130. doi:10.1016/j. 132. Jain A, Vale RD (2017) RNA phase transitions in repeat expansion disorders. ajhg.2011.05.015 Nature 546:243–247. doi:10.1038/nature22386 154. Kochetov AV, Palyanov A, Titov II, Grigorovich D, Sarai A, Kolchanov NA 133. Jansen A, Gemayel R, Verstrepen KJ (2012) Unstable microsatellite repeats (2007) AUG_hairpin: prediction of a downstream secondary structure facilitate rapid evolution of coding and regulatory sequences. Genome Dyn influencing the recognition of a translation start site. BMC Bioinformatics 7:108–125. doi:10.1159/000337121 8:318. doi:10.1186/1471-2105-8-318 134. Jiang J, Zhu Q, Gendron TF, Saberi S, McAlonis-Downes M, Seelman A, 155. Koide R, Ikeuchi T, Onodera O, Tanaka H, Igarashi S, Endo K, Takahashi H, Stauffer JE, Jafar-Nejad P, Drenner K, Schulte D et al (2016) Gain of Toxicity Kondo R, Ishikawa A, Hayashi T et al (1994) Unstable expansion of CAG from ALS/FTD-Linked Repeat Expansions in C9ORF72 Is Alleviated by repeat in hereditary dentatorubral-pallidoluysian atrophy (DRPLA). Nat Antisense Oligonucleotides Targeting GGGGCC-Containing RNAs. Neuron Genet 6:9–13. doi:10.1038/ng0194-9 90:535–550. doi:10.1016/j.neuron.2016.04.006 156. Koob MD, Moseley ML, Schut LJ, Benzow KA, Bird TD, Day JW, Ranum LP 135. Jokhi V, Ashley J, Nunnari J, Noma A, Ito N, Wakabayashi-Ito N, Moore MJ, (1999) An untranslated CTG expansion causes a novel form of Budnik V (2013) Torsin mediates primary envelopment of large spinocerebellar ataxia (SCA8). Nat Genet 21:379–384. doi:10.1038/7710 Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 18 of 22

157. Kota KP, Wagner SR, Huerta E, Underwood JM, Nickerson JA (2008) Binding 179. Li YY, Abu-Ghazalah R, Zamiri B, Macgregor RB Jr (2016) Concentration- of ATP to UAP56 is necessary for mRNA export. J Cell Sci 121:1526–1537. dependent conformational changes in GQ-forming ODNs. Biophys Chem doi:10.1242/jcs.021055 211:70–75. doi:10.1016/j.bpc.2016.02.002 158. Kozak M (1990) Downstream secondary structure facilitates recognition 180. Lin Y, Hubert L Jr, Wilson JH (2009) Transcription destabilizes triplet repeats. of initiator codons by eukaryotic . Proc Natl Acad Sci U S A Mol Carcinog 48:350–361. doi:10.1002/mc.20488 87:8301–8305 181. Lin Y, Leng M, Wan M, Wilson JH (2010) Convergent transcription through a 159. Kramer NJ, Carlomagno Y, Zhang YJ, Almeida S, Cook CN, Gendron TF, long CAG tract destabilizes repeats and induces apoptosis. Mol Cell Biol Prudencio M, Van Blitterswijk M, Belzil V, Couthouis J et al (2016) Spt4 30:4435–4451. doi:10.1128/MCB.00332-10 selectively regulates the expression of C9orf72 sense and antisense mutant 182. Lin Y, Mori E, Kato M, Xiang S, Wu L, Kwon I, McKnight SL (2016) Toxic PR transcripts. Science 353:708–712. doi:10.1126/science.aaf7791 Poly-Dipeptides Encoded by the C9orf72 Repeat Expansion Target LC 160. Krauss S, Griesche N, Jastrzebska E, Chen C, Rutschow D, Achmuller C, Dorn S, Domain Polymers. Cell 167:789–802 e712. doi:10.1016/j.cell.2016.10.003 Boesch SM, Lalowski M, Wanker E et al (2013) Translation of HTT mRNA with 183. Lin Y, Wilson JH (2012) Nucleotide excision repair, mismatch repair, and R- expanded CAG repeats is regulated by the MID1-PP2A protein complex. Nat loops modulate convergent transcription-induced cell death and repeat Commun 4:1511. doi:10.1038/ncomms2514 instability. PLoS One 7:e46807. doi:10.1371/journal.pone.0046807 161. Kremer EJ, Yu S, Pritchard M, Nagaraja R, Heitz D, Lynch M, Baker E, 184. Liquori CL, Ricker K, Moseley ML, Jacobsen JF, Kress W, Naylor SL, Day JW, Hyland VJ, Little RD, Wada Met al (1991) Isolation of a human DNA Ranum LP (2001) Myotonic dystrophy type 2 caused by a CCTG expansion sequence which spans the fragile X. Am J Hum Genet 49:656–661 in intron 1 of ZNF9. Science 293:864–867. doi:10.1126/science.1062125 162. Kumari D, Biacsi RE, Usdin K (2011) Repeat expansion affects both 185. Liu B, Steitz TA (2017) Structural insights into NusG regulating transcription transcription initiation and elongation in friedreich ataxia cells. J Biol Chem elongation. Nucleic Acids Res 45:968–974. doi:10.1093/nar/gkw1159 286:4209–4215. doi:10.1074/jbc.M110.194035 186. Liu CR, Chang CR, Chern Y, Wang TH, Hsieh WC, Shen WC, Chang CY, 163. Kurosaki T, Ueda S, Ishida T, Abe K, Ohno K, Matsuura T (2012) The unstable Chu IC, Deng N, Cohen SN et al (2012) Spt4 is selectively required for CCTG repeat responsible for myotonic dystrophy type 2 originates from an transcription of extended trinucleotide repeats. Cell 148:690–701. AluSx element into an early primate genome. PLoS One 7:e38379. doi:10.1016/j.cell.2011.12.032 doi:10.1371/journal.pone.0038379 187. Liu G, Chen X, Bissler JJ, Sinden RR, Leffak M (2010) Replication-dependent 164. Kwok CK, Marsico G, Sahakyan AB, Chambers VS, Balasubramanian S (2016) instability at (CTG) x (CAG) repeat hairpins in human cells. Nat Chem Biol rG4-seq reveals widespread formation of G-quadruplex structures in the 6:652–659. doi:10.1038/nchembio.416 human . Nat Methods 13:841–844. doi:10.1038/nmeth.3965 188. Liu J, Hu J, Ludlow AT, Pham JT, Shay JW, Rothstein JD, Corey DR 165. Kwon I, Xiang S, Kato M, Wu L, Theodoropoulous P, Wang T, Kim J, (2017) c9orf72 Disease-Related Foci Are Each Composed of One Mutant Yun J, Xie Y, McKnight SL (2014) Poly-dipeptides encoded by the Expanded Repeat RNA. Cell Chem Biol 24:141–148. doi:10.1016/j. C9orf72 repeats bind nucleoli, impede RNA biogenesis, and kill cells. chembiol.2016.12.018 Science 345:1139–1145 189. Llorente B, Smith CE, Symington LS (2008) Break-induced replication: what 166. La Spada AR, Taylor JP (2010) Repeat expansion disease: progress and is it and what is it for? Cell Cycle 7:859–864. doi:10.4161/cc.7.7.5613 puzzles in disease pathogenesis. Nat Rev Genet 11:247–258. doi:10.1038/ 190. Lokanga RA, Senejani AG, Sweasy JB, Usdin K (2015) Heterozygosity for a nrg2748 hypomorphic Polbeta mutation reduces the expansion frequency in a 167. La Spada AR, Wilson EM, Lubahn DB, Harding AE, Fischbeck KH (1991) mouse model of the Fragile X-related disorders. PLoS Genet 11:e1005181. Androgen receptor gene mutations in X-linked spinal and bulbar muscular doi:10.1371/journal.pgen.1005181 atrophy. Nature 352:77–79. doi:10.1038/352077a0 191. Lujan SA, Clausen AR, Clark AB, MacAlpine HK, MacAlpine DM, Malc EP, 168. Labbadia J, Morimoto RI (2013) Huntington's disease: underlying molecular Mieczkowski PA, Burkholder AB, Fargo DC, Gordenin DA et al (2014) mechanisms and emerging concepts. Trends Biochem Sci 38:378–385. Heterogeneous polymerase fidelity and mismatch repair bias genome doi:10.1016/j.tibs.2013.05.003 variation and composition. Genome Res 24:1751–1764. doi:10.1101/gr. 169. Lagier-Tourenne C, Baughn M, Rigo F, Sun S, Liu P, Li HR, Jiang J, Watt AT, 178335.114 Chun S, Katz M et al (2013) Targeted degradation of sense and antisense 192. MacDonald ME, Barnes G, Srinidhi J, Duyao MP, Ambrose CM, Myers RH, C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proc Gray J, Conneally PM, Young A, Penney J et al (1993) Gametic but not Natl Acad Sci U S A 110:E4530–E4539. doi:10.1073/pnas.1318835110 somatic instability of CAG repeat length in Huntington's disease. J Med 170. Lalioti MD, Scott HS, Buresi C, Rossier C, Bottani A, Morris MA, Malafosse A, Genet 30:982–986 Antonarakis SE (1997) Dodecamer repeat expansion in cystatin B gene in 193. Mahadevan M, Tsilfidis C, Sabourin L, Shutler G, Amemiya C, Jansen G, progressive myoclonus epilepsy. Nature 386:847–851. doi:10.1038/386847a0 Neville C, Narang M, Barcelo J, O'Hoy K et al (1992) Myotonic dystrophy 171. Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, mutation: an unstable CTG repeat in the 3' untranslated region of the gene. Dewar K, Doyle M, FitzHugh W et al (2001) Initial sequencing and analysis of Science 255:1253–1255 the human genome. Nature 409:860–921. doi:10.1038/35057062 194. Malgowska M, Gudanis D, Kierzek R, Wyszko E, Gabelica V, Gdaniec Z (2014) 172. Le Hir H, Izaurralde E, Maquat LE, Moore MJ (2000) The spliceosome Distinctive structural motifs of RNA G-quadruplexes composed of AGG, deposits multiple proteins 20-24 nucleotides upstream of mRNA exon-exon CGG and UGG trinucleotide repeats. Nucleic Acids Res 42:10196–10207. junctions. EMBO J 19:6860–6869. doi:10.1093/emboj/19.24.6860 doi:10.1093/nar/gku710 173. Lee JE, Cooper TA (2009) Pathogenic mechanisms of myotonic dystrophy. 195. Malkova A, Haber JE (2012) Mutations arising during repair of Biochem Soc Trans 37:1281–1286. doi:10.1042/BST0371281 chromosome breaks. Annu Rev Genet 46:455–473. doi:10.1146/annurev- 174. Lee KH, Zhang P, Kim HJ, Mitrea DM, Sarkar M, Freibaum BD, Cika J, genet-110711-155547 Coughlin M, Messing J, Molliex A et al (2016) C9orf72 Dipeptide Repeats 196. Mankodi A, Logigian E, Callahan L, McClain C, White R, Henderson D, Impair the Assembly, Dynamics, and Function of Membrane-Less Krym M, Thornton CA (2000) Myotonic dystrophy in transgenic mice Organelles. Cell 167(774-788):e717. doi:10.1016/j.cell.2016.10.002 expressing an expanded CUG repeat. Science 289:1769–1773 175. Lee YB, Chen HJ, Peres JN, Gomez-Deza J, Attig J, Stalekar M, Troakes C, 197. Mankodi A, Urbinati CR, Yuan QP, Moxley RT, Sansone V, Krym M, Nishimura AL, Scotter EL, Vance C et al (2013) Hexanucleotide Repeats in Henderson D, Schalling M, Swanson MS, Thornton CA (2001) Muscleblind ALS/FTD Form Length-Dependent RNA Foci, Sequester RNA Binding localizes to nuclear foci of aberrant RNA in myotonic dystrophy types 1 and Proteins, and Are Neurotoxic. Cell Rep 5:1178–1186. doi:10.1016/j.celrep. 2. Hum Mol Genet 10:2165–2170 2013.10.049 198. Mao YS, Zhang B, Spector DL (2011) Biogenesis and function of nuclear 176. Leffak M (2017) Break-induced replication links microsatellite expansion to bodies. Trends Genet 27:295–306. doi:10.1016/j.tig.2011.05.006 complex genome rearrangements. Bioessays 39. doi:10.1002/bies.201700025 199. Maquat LE, Li X (2001) Mammalian heat shock p70 and histone H4 177. Li L, Matsui M, Corey DR (2016) Activating frataxin expression by repeat- transcripts, which derive from naturally intronless genes, are immune to targeted nucleic acids. Nat Commun 7:10606. doi:10.1038/ncomms10606 nonsense-mediated decay. RNA 7:445–456 178. Li Y, Bor YC, Misawa Y, Xue Y, Rekosh D, Hammarskjold ML (2006) An intron 200. Maric M, Shao J, Ryan RJ, Wong CS, Gonzalez-Alegre P, Roller RJ (2011) A with a constitutive transport element is retained in a Tap messenger RNA. functional role for TorsinA in herpes simplex virus 1 nuclear egress. J Virol Nature 443:234–237. doi:10.1038/nature05107 85:9667–9679. doi:10.1128/JVI.05314-11 Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 19 of 22

201. Martinez-Rucobo FW, Sainsbury S, Cheung AC, Cramer P (2011) Architecture 221. Muranyi W, Haas J, Wagner M, Krohne G, Koszinowski UH (2002) of the RNA polymerase-Spt4/5 complex and basis of universal transcription Cytomegalovirus recruitment of cellular to dissolve the nuclear processivity. EMBO J 30:1302–1310. doi:10.1038/emboj.2011.64 lamina. Science 297:854–857. doi:10.1126/science.1071506 202. Mason SW, Greenblatt J (1991) Assembly of transcription elongation 222. Myers SJ, Huang Y, Genetta T, Dingledine R (2004) Inhibition of glutamate complexes containing the N protein of phage lambda and the Escherichia receptor 2 translation by a polymorphic repeat sequence in the 5'- coli elongation factors NusA, NusB, NusG, and S10. Genes Dev 5:1504–1512 untranslated leaders. J Neurosci 24:3489–3499. doi:10.1523/JNEUROSCI.4127- 203. Massenet S, Bertrand E, Verheggen C (2016) Assembly and trafficking of box 03.2004 C/D and H/ACA snoRNPs. RNA Biol 1–13. doi:10.1080/15476286.2016. 223. Nakamori M, Pearson CE, Thornton CA (2011) Bidirectional transcription 1243646 stimulates expansion and contraction of expanded (CTG)*(CAG) repeats. 204. Masuda S, Das R, Cheng H, Hurt E, Dorman N, Reed R (2005) Recruitment of Hum Mol Genet 20:580–588. doi:10.1093/hmg/ddq501 the human TREX complex to mRNA during splicing. Genes Dev 19:1512–1517. 224. Nakamura K, Jeong SY, Uchihara T, Anno M, Nagashima K, Nagashima T, doi:10.1101/gad.1302205 Ikeda S, Tsuji S, Kanazawa I (2001) SCA17, a novel autosomal dominant 205. Matsuura T, Yamagata T, Burgess DL, Rasmussen A, Grewal RP, Watase K, cerebellar ataxia caused by an expanded polyglutamine in TATA-binding Khajavi M, McCall AE, Davis CF, Zu L et al (2000) Large expansion of the protein. Hum Mol Genet 10:1441–1448 ATTCT pentanucleotide repeat in spinocerebellar ataxia type 10. Nat Genet 225. Neil AJ, Kim JC, Mirkin SM (2017) Precarious maintenance of simple DNA 26:191–194. doi:10.1038/79911 repeats in . BioEssays. doi:10.1002/bies.201700077 206. McMurray CT (2010) Mechanisms of trinucleotide repeat instability during 226. Nguyen L, Luu LM, Peng S, Serrano JF, Chan HY, Zimmerman SC (2015) human development. Nat Rev Genet 11:786–799. doi:10.1038/nrg2828 Rationally Designed Small Molecules That Target Both the DNA and RNA 207. Meng F, Na I, Kurgan L, Uversky VN (2015) Compartmentalization and Causing Myotonic Dystrophy Type 1. J Am Chem Soc 137:14180–14189. Functionality of Nuclear Disorder: Intrinsic Disorder and Protein-Protein doi:10.1021/jacs.5b09266 Interactions in Intra-Nuclear Compartments. Int J Mol Sci 17. doi:10.3390/ 227. Niblock M, Smith BN, Lee YB, Sardone V, Topp S, Troakes C, Al-Sarraj S, ijms17010024 Leblond CS, Dion PA, Rouleau GA et al (2016) Retention of hexanucleotide 208. Meservy JL, Sargent RG, Iyer RR, Chan F, McKenzie GJ, Wells RD, Wilson JH repeat-containing intron in C9orf72 mRNA: implications for the (2003) Long CTG tracts from the myotonic dystrophy gene induce deletions pathogenesis of ALS/FTD. Acta neuropathol Commun 4:18. doi:10.1186/ and rearrangements during recombination at the APRT locus in CHO cells. s40478-016-0289-4 Mol Cell Biol 23:3152–3162 228. Nunes VS, Moretti NS (2017) Nuclear subcompartments: an overview. Cell 209. Metsu S, Rainger JK, Debacker K, Bernhard B, Rooms L, Grafodatskaya D, Biol Int 41:2–7. doi:10.1002/cbin.10703 Weksberg R, Fombonne E, Taylor MS, Scherer SW et al (2014) A CGG-repeat 229. O'Geen H, Yu AS, Segal DJ (2015) How specific is CRISPR/Cas9 really? Curr expansion mutation in ZNF713 causes FRA7A: association with autistic Opin Chem Biol 29:72–78. doi:10.1016/j.cbpa.2015.10.001 spectrum disorder in two families. Hum Mutat 35:1295–1300. doi:10.1002/ 230. Oberle I, Rousseau F, Heitz D, Kretz C, Devys D, Hanauer A, Boue J, humu.22683 Bertheas MF, Mandel JL (1991) Instability of a 550- DNA segment and 210. Metsu S, Rooms L, Rainger J, Taylor MS, Bengani H, Wilson DI, Chilamakuri CS, abnormal methylation in fragile X syndrome. Science 252:1097–1102 Morrison H, Vandeweyer G, Reyniers E et al (2014) FRA2A is a CGG repeat 231. Ohshima K, Montermini L, Wells RD, Pandolfo M (1998) Inhibitory effects of expansion associated with silencing of AFF3. PLoS Genet 10:e1004242. expanded GAA.TTC triplet repeats from intron I of the Friedreich ataxia doi:10.1371/journal.pgen.1004242 gene on transcription and replication in vivo. J Biol Chem 273:14588–14595 211. Michael TP, Park S, Kim TS, Booth J, Byer A, Sun Q, Chory J, Lee K (2007) 232. Orr HT, Chung MY, Banfi S, Kwiatkowski TJ Jr, Servadio A, Beaudet AL, Simple sequence repeats provide a substrate for phenotypic variation in the McCall AE, Duvick LA, Ranum LP, Zoghbi HY (1993) Expansion of an Neurospora crassa circadian clock. PLoS One 2:e795. doi:10.1371/journal. unstable trinucleotide CAG repeat in spinocerebellar ataxia type 1. Nat pone.0000795 Genet 4:221–226. doi:10.1038/ng0793-221 212. Mirkin SM (2007) Expandable DNA repeats and human disease. Nature 233. Ozdilek BA, Thompson VF, Ahmed NS, White CI, Batey RT, Schwartz JC 447:932–940. doi:10.1038/nature05977 (2017) Intrinsically disordered RGG/RG domains mediate degenerate 213. Mizielinska S, Gronke S, Niccoli T, Ridler CE, Clayton EL, Devoy A, Moens T, specificity in RNA binding. Nucleic Acids Res. doi:10.1093/nar/gkx460 Norona FE, Woollacott IO, Pietrzyk J et al (2014) C9orf72 repeat expansions 234. Park H, Gonzalez AL, Yildirim I, Tran T, Lohman JR, Fang P, Guo M, cause neurodegeneration in Drosophila through arginine-rich proteins. Disney MD (2015) Crystallographic and Computational Analyses of Science 345:1192–1194. doi:10.1126/science.1256800 AUUCU Repeating RNA That Causes Spinocerebellar Ataxia Type 10 214. Mohan A, Goodwin M, Swanson MS (2014) RNA-protein interactions in (SCA10). Biochemistry 54:3851–3859. doi:10.1021/acs.biochem.5b00551 unstable microsatellite diseases. Brain Res 1584:3–14. doi:10.1016/j.brainres. 235. Paul S, Dansithong W, Jog SP, Holt I, Mittal S, Brook JD, Morris GE, Comai L, 2014.03.039 Reddy S (2011) Expanded CUG repeats Dysregulate RNA splicing by 215. Monteys AM, Ebanks SA, Keiser MS, Davidson BL (2017) CRISPR/Cas9 Editing altering the stoichiometry of the muscleblind 1 complex. J Biol Chem of the Mutant Huntingtin Allele In Vitro and In Vivo. Mol Ther 25:12–23. 286:38427–38438. doi:10.1074/jbc.M111.255224 doi:10.1016/j.ymthe.2016.11.010 236. Payseur BA, Jing P, Haasl RJ (2011) A genomic portrait of human microsatellite 216. Mori K, Arzberger T, Grasser FA, Gijselinck I, May S, Rentzsch K, Weng SM, variation. Mol Biol Evol 28:303–312. doi:10.1093/molbev/msq198 Schludi MH, van der Zee J, Cruts M et al (2013) Bidirectional transcripts of 237. Pearson CE, Sinden RR (1996) Alternative structures in duplex DNA formed the expanded C9orf72 hexanucleotide repeat are translated into within the trinucleotide repeats of the myotonic dystrophy and fragile X aggregating dipeptide repeat proteins. Acta Neuropathol 126:881–893. loci. Biochemistry 35:5041–5053. doi:10.1021/bi9601013 doi:10.1007/s00401-013-1189-3 238. Peng R, Lin G, Li J (2015) Potential Pitfalls of CRISPR/Cas9-mediated Genome 217. Mori K, Lammich S, Mackenzie IR, Forne I, Zilow S, Kretzschmar H, Editing. The FEBS J. doi:10.1111/febs.13586 Edbauer D, Janssens J, Kleinberger G, Cruts M et al (2013) hnRNP A3 239. Pfister EL, Kennington L, Straubhaar J, Wagh S, Liu W, DiFiglia M, binds to GGGGCC repeats and is a constituent of p62-positive/TDP43- Landwehrmeyer B, Vonsattel JP, Zamore PD, Aronin N (2009) Five siRNAs negative inclusions in the hippocampus of patients with C9orf72 targeting three SNPs may provide therapy for three-quarters of Huntington's mutations. Acta Neuropathol 125:413–423. doi:10.1007/s00401-013-1088-7 disease patients. Curr Biol 19:774–778. doi:10.1016/j.cub.2009.03.030 218. Mori K, Weng SM, Arzberger T, May S, Rentzsch K, Kremmer E, Schmid B, 240. Pineda M, Moghadam F, Ebrahimkhani MR, Kiani S (2017) Engineered Kretzschmar HA, Cruts M, Van Broeckhoven C et al (2013) The C9orf72 CRISPR Systems for Next Generation Gene Therapies. ACS Synthetic Biol. GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in doi:10.1021/acssynbio.7b00011 FTLD/ALS. Science 339:1335–1338. doi:10.1126/science.1232927 241. Pluciennik A, Burdett V, Baitinger C, Iyer RR, Shi K, Modrich P (2013) 219. Moussavi-Baygi R, Mofrad MR (2016) Rapid Brownian Motion Primes Extrahelical (CAG)/(CTG) triplet repeat elements support proliferating cell Ultrafast Reconstruction of Intrinsically Disordered Phe-Gly Repeats Inside nuclear antigen loading and MutLalpha activation. Proc Natl the Nuclear Pore Complex. Sci Rep 6:29991. doi:10.1038/srep29991 Acad Sci U S A 110:12277–12282. doi:10.1073/pnas.1311325110 220. Muhlemann O, Mock-Casagrande CS, Wang J, Li S, Custodio N, Carmo- 242. Polleys EJ, NCM H, Freudenreich CH (2017) Role of recombination and Fonseca M, Wilkinson MF, Moore MJ (2001) Precursor RNAs harboring replication fork restart in repeat instability. DNA Repair 56:156–165. nonsense codons accumulate near the site of transcription. Mol Cell 8:33–43 doi:10.1016/j.dnarep.2017.06.018 Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 20 of 22

243. Querido E, Gallardo F, Beaudoin M, Menard C, Chartrand P (2011) Stochastic repeat expansion block nuclear import and export. Proc Natl Acad Sci and reversible aggregation of mRNA with expanded CUG-triplet repeats. USA114:E1111–E1117. doi:10.1073/pnas.1620293114 J Cell Sci 124:1703–1714 Doi 2461242/jcs.073270 265. Shin JW, Kim KH, Chao MJ, Atwal RS, Gillis T, MacDonald ME, Gusella JF, 244. Raca G, Siyanova EY, McMurray CT, Mirkin SM (2000) Expansion of the Lee JM (2016) Permanent inactivation of Huntington's disease mutation (CTG)(n) repeat in the 5'-UTR of a reporter gene impedes translation. by personalized allele-specific CRISPR/Cas9. Hum Mol Genet 25:4566–4576. Nucleic Acids Res 28:3943–3949 doi:10.1093/hmg/ddw286 245. Ranum LP, Cooper TA (2006) RNA-mediated neuromuscular disorders. Annu 266. Shishkin AA, Voineagu I, Matera R, Cherng N, Chernet BT, Krasilnikova MM, Rev Neurosci 29:259–277. doi:10.1146/annurev.neuro.29.051605.113014 Narayanan V, Lobachev KS, Mirkin SM (2009) Large-scale expansions of 246. Reddy K, Schmidt MH, Geist JM, Thakkar NP, Panigrahi GB, Wang YH, Friedreich's ataxia GAA repeats in yeast. Mol Cell 35:82–92. doi:10.1016/j. Pearson CE (2014) Processing of double-R-loops in (CAG).(CTG) and molcel.2009.06.017 C9orf72 (GGGGCC).(GGCCCC) repeats causes instability. Nucleic Acids 267. Shoemaker CJ, Green R (2012) Translation drives mRNA quality control. Nat Res 42:10473–10487. doi:10.1093/nar/gku658 Struct Mol Biol 19:594–601. doi:10.1038/nsmb.2301 247. Reddy K, Zamiri B, Stanley SYR, Macgregor RB, Pearson CE (2013) The 268. Shoubridge C, Gecz J (2012) Polyalanine tract disorders and neurocognitive Disease-associated r(GGGGCC)(n) Repeat from the C9orf72 Gene Forms . Adv Exp Med Biol 769:185–203 Tract Length-dependent Uni- and Multimolecular RNA G-quadruplex 269. Shukla S, Parker R (2016) Hypo- and Hyper-Assembly Diseases of RNA-Protein Structures. J Biol Chem 288:9860–9866. doi:10.1074/jbc.C113.452532 Complexes. Trends Mol Med 22:615–628. doi:10.1016/j.molmed.2016.05.005 248. Renton AE, Majounie E, Waite A, Simon-Sanchez J, Rollinson S, Gibbs JR, 270. Skourti-Stathaki K, Proudfoot NJ, Gromak N (2011) Human senataxin Schymick JC, Laaksovirta H, van Swieten JC, Myllykangas L et al (2011) A resolves RNA/DNA hybrids formed at transcriptional pause sites to Hexanucleotide Repeat Expansion in C9ORF72 Is the Cause of Chromosome promote Xrn2-dependent termination. Mol Cell 42:794–805. doi:10.1016/ 9p21-Linked ALS-FTD. Neuron 72:257–268. doi:10.1016/j.neuron.2011.09.010 j.molcel.2011.04.026 249. Richard GF, Cyncynatus C, Dujon B (2003) Contractions and expansions of 271. Slean MM, Panigrahi GB, Castel AL, Pearson AB, Tomkinson AE, Pearson CE CAG/CTG trinucleotide repeats occur during ectopic in (2016) Absence of MutSbeta leads to the formation of slipped-DNA for yeast, by a MUS81-independent mechanism. J Mol Biol 326:769–782 CTG/CAG contractions at primate replication forks. DNA repair 42:107–118. 250. Richard GF, Goellner GM, McMurray CT, Haber JE (2000) Recombination- doi:10.1016/j.dnarep.2016.04.002 induced CAG trinucleotide repeat expansions in yeast involve the MRE11- 272. Sleeman JE, Trinkle-Mulcahy L (2014) Nuclear bodies: new insights into RAD50-XRS2 complex. EMBO J 19:2381–2390. doi:10.1093/emboj/19.10.2381 assembly/dynamics and disease relevance. Curr Opin Cell Biol 28:76–83. 251. Richard P, Manley JL (2016) R Loops and Links to Human Disease. J Mol doi:10.1016/j.ceb.2014.03.004 Biol. doi:10.1016/j.jmb.2016.08.031 273. Sloan KE, Gleizes PE, Bohnsack MT (2016) Nucleocytoplasmic Transport of 252. Rzuczek SG, Colgan LA, Nakai Y, Cameron MD, Furling D, Yasuda R, Disney MD RNAs and RNA-Protein Complexes. J Mol Biol 428:2040–2059. doi:10.1016/j. (2017) Precise small-molecule recognition of a toxic CUG RNA repeat jmb.2015.09.023 expansion. Nat Chem Biol 13:188–193. doi:10.1038/nchembio.2251 274. Smith KP, Byron M, Johnson C, Xing Y, Lawrence JB (2007) Defining early 253. Sanpei K, Takano H, Igarashi S, Sato T, Oyake M, Sasaki H, Wakisaka A, steps in mRNA transport: mutant mRNA in myotonic dystrophy type I is Tashiro K, Ishida Y, Ikeuchi T et al (1996) Identification of the spinocerebellar blocked at entry into SC-35 domains. J Cell Biol 178:951–964. doi:10.1083/ ataxia type 2 gene using a direct identification of repeat expansion and jcb.200706048 cloning technique, DIRECT. Nat Genet 14:277–284. doi:10.1038/ng1196-277 275. Sobczak K, Wheeler TM, Wang W, Thornton CA (2013) RNA interference 254. Sanz LA, Hartono SR, Lim YW, Steyaert S, Rajpurkar A, Ginno PA, Xu X, targeting CUG repeats in a mouse model of myotonic dystrophy. Mol Ther Chedin F (2016) Prevalent, Dynamic, and Conserved R-Loop Structures 21:380–387. doi:10.1038/mt.2012.222 Associate with Specific Epigenomic Signatures in Mammals. Mol Cell 276. Sofola OA, Jin P, Qin Y, Duan R, Liu H, de Haro M, Nelson DL, Botas J (2007) 63:167–178. doi:10.1016/j.molcel.2016.05.032 RNA-binding proteins hnRNP A2/B1 and CUGBP1 suppress fragile X CGG 255. Sathasivam K, Neueder A, Gipson TA, Landles C, Benjamin AC, Bondulich MK, premutation repeat-induced neurodegeneration in a Drosophila model of Smith DL, Faull RL, Roos RA, Howland D et al (2013) Aberrant splicing of HTT FXTAS. Neuron 55:565–571. doi:10.1016/j.neuron.2007.07.021 generates the pathogenic exon 1 protein in Huntington disease. Proc Natl 277. Speese SD, Ashley J, Jokhi V, Nunnari J, Barria R, Li Y, Ataman B, Koon A, Acad Sci U S A 110:2366–2370. doi:10.1073/pnas.1221891110 Chang YT, Li Q et al (2012) Nuclear envelope budding enables large 256. Sato N, Amino T, Kobayashi K, Asakawa S, Ishiguro T, Tsunemi T, Takahashi M, ribonucleoprotein particle export during synaptic Wnt signaling. Cell Matsuura T, Flanigan KM, Iwasaki S et al (2009) Spinocerebellar ataxia type 31 is 149:832–846. doi:10.1016/j.cell.2012.03.032 associated with "inserted" penta-nucleotide repeats containing (TGGAA)n. Am 278. Spiro C, Pelletier R, Rolfsmeier ML, Dixon MJ, Lahue RS, Gupta G, Park MS, J Hum Genet 85:544–557. doi:10.1016/j.ajhg.2009.09.019 Chen X, Mariappan SV, McMurray CT (1999) Inhibition of FEN-1 processing 257. Sawaya S, Bagshaw A, Buschiazzo E, Kumar P, Chowdhury S, Black MA, by DNA secondary structure at trinucleotide repeats. Mol Cell 4:1079–1085 Gemmell N (2013) Microsatellite tandem repeats are abundant in human 279. Stern A, Brown M, Nickel P, Meyer TF (1986) Opacity genes in Neisseria promoters and are associated with regulatory elements. PLoS One 8:e54710. gonorrhoeae: control of phase and antigenic variation. Cell 47:61–71 doi:10.1371/journal.pone.0054710 280. Strack RL, Disney MD, Jaffrey SR (2013) A superfolding Spinach2 reveals the 258. Sawyer LA, Hennessy JM, Peixoto AA, Rosato E, Parkinson H, Costa R, dynamic nature of trinucleotide repeat-containing RNA. Nat Methods Kyriacou CP (1997) Natural variation in a Drosophila clock gene and 10:1219–1224. doi:10.1038/Nmeth.2701 temperature compensation. Science 278:2117–2120 281. Su Z, Zhang Y, Gendron TF, Bauer PO, Chew J, Yang WY, Fostvedt E, 259. Schmidt K, Butler JS (2013) Nuclear RNA surveillance: role of TRAMP in Jansen-West K, Belzil VV, Desaro P et al (2014) Discovery of a controlling exosome specificity. Wiley Interdiscip Rev RNA 4:217–231. and lead small molecules to target r(GGGGCC)-associated defects in doi:10.1002/wrna.1155 c9FTD/ALS. Neuron 83:1043–1050. doi:10.1016/j.neuron.2014.07.041 260. Schmidt MH, Pearson CE (2016) Disease-associated repeat instability and 282. Sun X, Li PP, Zhu S, Cohen R, Marque LO, Ross CA, Pulst SM, Chan HY, mismatch repair. DNA Repair 38:117–126. doi:10.1016/j.dnarep.2015.11.008 Margolis RL, Rudnicki DD (2015) Nuclear retention of full-length HTT 261. Scoles DR, Ho MH, Dansithong W, Pflieger LT, Petersen LW, Thai KK, Pulst SM RNA is mediated by splicing factors MBNL1 and U2AF65. Sci Rep (2015) Repeat Associated Non-AUG Translation (RAN Translation) Dependent 5:12521. doi:10.1038/srep12521 on Sequence Downstream of the ATXN2 CAG Repeat. PLoS One 10:e0128769. 283. Sundararajan R, Gellon L, Zunder RM, Freudenreich CH (2010) Double-strand doi:10.1371/journal.pone.0128769 break repair pathways protect against CAG/CTG repeat expansions, 262. Sellier C, Rau F, Liu Y, Tassone F, Hukema RK, Gattoni R, Schneider A, contractions and repeat-mediated chromosomal fragility in Saccharomyces Richard S, Willemsen R, Elliott DJ et al (2010) Sam68 sequestration and cerevisiae. Genetics 184:65–77. doi:10.1534/genetics.109.111039 partial loss of function are associated with splicing alterations in FXTAS 284. Supek F, Lehner B (2015) Differential DNA mismatch repair underlies patients. EMBO J 29:1248–1261. doi:10.1038/emboj.2010.21 mutation rate variation across the human genome. Nature 521:81–84. 263. Shepard S, McCreary M, Fedorov A (2009) The peculiarities of large intron doi:10.1038/nature14173 splicing in animals. PLoS One 4:e7853. doi:10.1371/journal.pone.0007853 285. Sutcliffe JS, Nelson DL, Zhang F, Pieretti M, Caskey CT, Saxe D, Warren ST 264. Shi KY, Mori E, Nizami ZF, Lin Y, Kato M, Xiang S, Wu LC, Ding M, Yu Y, (1992) DNA methylation represses FMR-1 transcription in fragile X Gall JG et al (2017) Toxic PRn poly-dipeptides encoded by the C9orf72 syndrome. Hum Mol Genet 1:397–400 Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 21 of 22

286. Tao Z, Wang H, Xia Q, Li K, Jiang X, Xu G, Wang G, Ying Z (2015) Nucleolar the hSpt5 dimerization interface. Biochem J 425:373–380. doi:10.1042/ stress and impaired stress granule formation contribute to C9orf72 RAN BJ20091422 translation-induced cytotoxicity. Hum Mol Genet. doi:10.1093/hmg/ddv005 309. Werner F (2012) A nexus for gene expression-molecular mechanisms of 287. Tassone F, Hagerman RJ, Taylor AK, Hagerman PJ (2001) A majority of fragile Spt5 and NusG in the three domains of life. J Mol Biol 417:13–27. X males with methylated, full mutation have significant levels of doi:10.1016/j.jmb.2012.01.031 FMR1 messenger RNA. J Med Genet 38:453–456 310. Wheeler TM, Sobczak K, Lueck JD, Osborne RJ, Lin X, Dirksen RT, Thornton CA 288. Taylor JP, Brown RH Jr, Cleveland DW (2016) Decoding ALS: from genes to (2009) Reversal of RNA by displacement of protein sequestered on mechanism. Nature 539:197–206. doi:10.1038/nature20413 triplet repeat RNA. Science 325:336–339 289. Thandapani P, O'Connor TR, Bailey TL, Richard S (2013) Defining the RGG/RG 311. White MC, Gao R, Xu W, Mandal SM, Lim JG, Hazra TK, Wakamiya M, motif. Mol Cell 50:613–623. doi:10.1016/j.molcel.2013.05.021 Edwards SF, Raskin S, Teive HA et al (2010) Inactivation of hnRNP K by 290. Todd PK, Oh SY, Krans A, He F, Sellier C, Frazer M, Renoux AJ, Chen KC, expanded intronic AUUCU repeat induces apoptosis via translocation of Scaglione KM, Basrur V et al (2013) CGG repeat-associated translation PKCdelta to mitochondria in spinocerebellar ataxia 10. PLoS Genet mediates neurodegeneration in fragile X tremor ataxia syndrome. Neuron 6:e1000984. doi:10.1371/journal.pgen.1000984 78:440–455. doi:10.1016/j.neuron.2013.03.026 312. Wilburn B, Rudnicki DD, Zhao J, Weitz TM, Cheng Y, Gu X, Greiner E, 291. Toth G, Gaspari Z, Jurka J (2000) Microsatellites in different eukaryotic Park CS, Wang N, Sopher BL et al (2011) An antisense CAG repeat genomes: survey and analysis. Genome Res 10:967–981 transcript at JPH3 locus mediates expanded polyglutamine protein 292. Tran T, Childs-Disney JL, Liu B, Guan L, Rzuczek S, Disney MD (2014) toxicity in Huntington's disease-like 2 mice. Neuron 70:427–440. Targeting the r(CGG) repeats that cause FXTAS with modularly assembled doi:10.1016/j.neuron.2011.03.021 small molecules and oligonucleotides. Acs Chem Biol 9:904–912. 313. Wojciechowska M, Olejniczak M, Galka-Marciniak P, Jazurek M, Krzyzosiak WJ doi:10.1021/cb400875u (2014) RAN translation and frameshifting as translational challenges at 293. Trinklein ND, Aldred SF, Hartman SJ, Schroeder DI, Otillar RP, Myers RM simple repeats of human neurodegenerative disorders. Nucleic Acids Res (2004) An abundance of bidirectional promoters in the human genome. 42:11849–11864. doi:10.1093/nar/gku794 Genome Res 14:62–66. doi:10.1101/gr.1982804 314. Wong CH, Fu Y, Ramisetty SR, Baranger AM, Zimmerman SC (2011) Selective 294. Turner EM, Brown RS, Laudermilch E, Tsai PL, Schlieker C (2015) The Torsin inhibition of MBNL1-CCUG interaction by small molecules toward potential Activator LULL1 Is Required for Efficient Growth of Herpes Simplex Virus 1. therapeutic agents for myotonic dystrophy type 2 (DM2). Nucleic Acids Res J Virol 89:8444–8452. doi:10.1128/JVI.01143-15 39:8881–8890. doi:10.1093/nar/gkr415 295. Urbanek MO, Jazurek M, Switonski PM, Figura G, Krzyzosiak WJ (2016) 315. Woodward LA, Mabin JW, Gangras P, Singh G (2016) The exon junction Nuclear speckles are detention centers for transcripts containing expanded complex: a lifelong guardian of mRNA fate. Wiley Int Rev RNA. doi:10.1002/ CAG repeats. Biochim Biophys Acta 1862:1513–1520. doi:10.1016/j.bbadis. wrna.1411 2016.05.015 316. Xi Z, Zhang M, Bruni AC, Maletta RG, Colao R, Fratta P, Polke JM, 296. Usdin K (2008) The biological effects of simple tandem repeats: lessons Sweeney MG, Mudanohwo E, Nacmias B et al (2015) The C9orf72 from the repeat expansion diseases. Genome Res 18:1011–1019. repeat expansion itself is methylated in ALS and FTLD patients. Acta doi:10.1101/gr.070409.107 18/7/1011 Neuropathol 129:715–727. doi:10.1007/s00401-015-1401-8 297. Usdin K, House NC, Freudenreich CH (2015) Repeat instability during DNA 317. Xi Z, Zinman L, Moreno D, Schymick J, Liang Y, Sato C, Zheng Y, Ghani M, repair: Insights from model systems. Crit Rev Biochem Mol Biol 50:142–167. Dib S, Keith J et al (2013) Hypermethylation of the CpG island near the doi:10.3109/10409238.2014.999192 G4C2 repeat in ALS with a C9orf72 expansion. Am J Hum Genet 92:981–989. 298. Usdin K, Kumari D (2015) Repeat-mediated epigenetic dysregulation of the doi:10.1016/j.ajhg.2013.04.017 FMR1 gene in the fragile X-related disorders. Front Genet 6:192. doi:10.3389/ 318. Xia G, Gao Y, Jin S, Subramony SH, Terada N, Ranum LP, Swanson MS, fgene.2015.00192 Ashizawa T (2015) Genome modification leads to reversal in 299. van Agtmaal EL, Andre LM, Willemse M, Cumming SA, van Kessel IDG, human myotonic dystrophy type 1 induced pluripotent stem cell-derived van den Broek W, Gourdon G, Furling D, Mouly V, Monckton DG et al (2017) neural stem cells. Stem Cells 33:1829–1838. doi:10.1002/stem.1970 CRISPR/Cas9-Induced (CTGCAG)n Repeat Instability in the Myotonic 319. Xu Z, Poidevin M, Li X, Li Y, Shu L, Nelson DL, Li H, Hales CM, Gearing M, Dystrophy Type 1 Locus: Implications for Therapeutic Genome Editing. Mol Wingo TS et al (2013) Expanded GGGGCC repeat RNA associated with Ther 25:24–43. doi:10.1016/j.ymthe.2016.10.014 amyotrophic lateral sclerosis and frontotemporal dementia causes 300. van Blitterswijk M, DeJesus-Hernandez M, Rademakers R (2012) How do neurodegeneration. Proc Natl Acad Sci U S A 110:7778–7783. doi:10.1073/ C9ORF72 repeat expansions cause amyotrophic lateral sclerosis and pnas.1219643110 frontotemporal dementia: can we learn from other noncoding repeat 320. Yamada NA, Smith GA, Castro A, Roques CN, Boyer JC, Farber RA (2002) expansion disorders? Curr Opin Neurol 25:689–700 Relative rates of insertion and deletion mutations in dinucleotide repeats of 301. Van Der Kelen K, Beyaert R, Inze D, De Veylder L (2009) Translational control various lengths in mismatch repair proficient mouse and mismatch repair of eukaryotic gene expression. Crit Rev Biochem Mol Biol 44:143–168. deficient human cells. Mutat Res 499:213–225 doi:10.1080/10409230902882090 321. Yamazaki T, Hirose T (2015) The building process of the functional 302. van Hoof A, Frischmeyer PA, Dietz HC, Parker R (2002) Exosome-mediated paraspeckle with long non-coding RNAs. Front Biosci (Elite Ed) 7:1–41 recognition and degradation of mRNAs lacking a termination codon. 322. Yang W, Tu Z, Sun Q, Li XJ (2016) CRISPR/Cas9: Implications for Modeling Science 295:2262–2264. doi:10.1126/science.1067272 and Therapy of Neurodegenerative Diseases. Front Mol Neurosci 9:30. 303. Verkerk AJ, Pieretti M, Sutcliffe JS, Fu YH, Kuhl DP, Pizzuti A, Reiner O, doi:10.3389/fnmol.2016.00030 Richards S, Victoria MF, Zhang FP et al (1991) Identification of a gene (FMR-1) 323. Yu K, Chedin F, Hsieh CL, Wilson TE, Lieber MR (2003) R-loops at containing a CGG repeat coincident with a breakpoint cluster region immunoglobulin class switch regions in the chromosomes of stimulated B exhibiting length variation in fragile X syndrome. Cell 65:905–914 cells. Nat Immunol 4:442–451. doi:10.1038/ni919 304. Villasante A, Abad JP, Mendez-Lago M (2007) were derived 324. Yu K, Roy D, Huang FT, Lieber MR (2006) Detection and structural analysis of from telomeres during the evolution of the eukaryotic chromosome. Proc R-loops. Methods Enzymol 409:316–329. doi:10.1016/S0076-6879(05)09018-X Natl Acad Sci U S A 104:10542–10547. doi:10.1073/pnas.0703808104 325. Zamiri B, Reddy K, Macgregor RB Jr, Pearson CE (2014) TMPyP4 porphyrin 305. Wada T, Takagi T, Yamaguchi Y, Ferdous A, Imai T, Hirose S, Sugimoto S, distorts RNA G-quadruplex structures of the disease-associated r(GGGGCC)n Yano K, Hartzog GA, Winston F et al (1998) DSIF, a novel transcription repeat of the C9orf72 gene and blocks interaction of RNA-binding proteins. elongation factor that regulates RNA polymerase II processivity, is J Biol Chem 289:4653–4659. doi:10.1074/jbc.C113.502336 composed of human Spt4 and Spt5 homologs. Genes Dev 12:343–356 326. Zhang K, Donnelly CJ, Haeusler AR, Grima JC, Machamer JB, Steinwald P, 306. Warren ST (1997) Polyalanine expansion in might result from Daley EL, Miller SJ, Cunningham KM, Vidensky S et al (2015) The C9orf72 unequal crossing-over of HOXD13. Science 275:408–409 repeat expansion disrupts nucleocytoplasmic transport. Nature 525:56–61. 307. Weiser JN, Love JM, Moxon ER (1989) The molecular mechanism of phase doi:10.1038/nature14973 variation of H. influenzae lipopolysaccharide. Cell 59:657–665 327. Zhang N, Ashizawa T (2017) RNA toxicity and foci formation in 308. Wenzel S, Martins BM, Rosch P, Wohrl BM (2009) Crystal structure of the microsatellite expansion diseases. Curr Opin Genet Dev 44:17–29. human transcription elongation factor DSIF hSpt4 subunit in complex with doi:10.1016/j.gde.2017.01.005 Rohilla and Gagnon Acta Neuropathologica Communications (2017) 5:63 Page 22 of 22

328. Zhang S, Binari R, Zhou R, Perrimon N (2010) A genomewide RNA interference screen for modifiers of aggregates formation by mutant Huntingtin in Drosophila. Genetics 184:1165–1179. doi:10.1534/genetics. 109.112516 329. Zhang Y, Roland C, Sagui C (2016) Structure and Dynamics of DNA and RNA Double Helices Obtained from the GGGGCC and CCCCGG Hexanucleotide Repeats That Are the Hallmark of C9FTD/ALS Diseases. ACS Chem Neurosci. doi:10.1021/acschemneuro.6b00348 330. Zhang Y, Shishkin AA, Nishida Y, Marcinkowski-Desmond D, Saini N, Volkov KV, Mirkin SM, Lobachev KS (2012) Genome-wide screen identifies pathways that govern GAA/TTC repeat fragility and expansions in dividing and nondividing yeast cells. Mol Cell 48:254–265. doi:10.1016/j.molcel.2012.08.002 331. Zhao J, Bacolla A, Wang G, Vasquez KM (2010) Non-B DNA structure- induced genetic instability and evolution. Cell Mol Life Sci 67:43–62. doi:10.1007/s00018-009-0131-2 332. Zhao T, Hong Y, Li XJ, Li SH (2016) Subcellular Clearance and Accumulation of Huntington Disease Protein: A Mini-Review. Front Mol Neurosci 9:27. doi:10.3389/fnmol.2016.00027 333. Zhao X, Usdin K (2015) The Repeat Expansion Diseases: The dark side of DNA repair. DNA Repair 32:96–105 334. Zhao XN, Lokanga R, Allette K, Gazy I, Wu D, Usdin K (2016) A MutSbeta- Dependent Contribution of MutSalpha to Repeat Expansions in Fragile X Premutation Mice? PLoS Genet 12:e1006190. doi:10.1371/journal.pgen. 1006190 335. Zhou Z, Luo MJ, Straesser K, Katahira J, Hurt E, Reed R (2000) The protein Aly links pre-messenger-RNA splicing to nuclear export in metazoans. Nature 407:401–405. doi:10.1038/35030160 336. Zhu L, Brangwynne CP (2015) Nuclear bodies: the emerging biophysics of nucleoplasmic phases. Curr Opin Cell Biol 34:23–30. doi:10.1016/j.ceb.2015. 04.003 337. Zhuchenko O, Bailey J, Bonnen P, Ashizawa T, Stockton DW, Amos C, Dobyns WB, Subramony SH, Zoghbi HY, Lee CC (1997) Autosomal dominant cerebellar ataxia (SCA6) associated with small polyglutamine expansions in the alpha 1A-voltage-dependent calcium channel. Nat Genet 15:62–69. doi:10.1038/ng0197-62 338. Zinder JC, Lima CD (2017) Targeting RNA for processing or destruction by the eukaryotic RNA exosome and its cofactors. Genes Dev 31:88–100. doi:10.1101/gad.294769.116 339. Zu T, Gibbens B, Doty NS, Gomes-Pereira M, Huguet A, Stone MD, Margolis J, Peterson M, Markowski TW, Ingram MA et al (2011) Non- ATG-initiated translation directed by microsatellite expansions. Proc Natl Acad Sci U S A 108:260–265. doi:10.1073/pnas.1013343108 340. Zu T, Liu Y, Banez-Coronel M, Reid T, Pletnikova O, Lewis J, Miller TM, Harms MB, Falchook AE, Subramony SH et al (2013) RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia. Proc Natl Acad Sci U S A 110:E4968–E4977. doi:10.1073/pnas. 1315438110 341. Zumwalt M, Ludwig A, Hagerman PJ, Dieckmann T (2007) Secondary structure and dynamics of the r(CGG) repeat in the mRNA of the fragile X mental retardation 1 (FMR1) gene. RNA Biol 4:93–100

Submit your next manuscript to BioMed Central and we will help you at every step:

• We accept pre-submission inquiries • Our selector tool helps you to find the most relevant journal • We provide round the clock customer support • Convenient online submission • Thorough peer review • Inclusion in PubMed and all major indexing services • Maximum visibility for your research

Submit your manuscript at www.biomedcentral.com/submit